US11579140B2 - Methods related to bronchial premalignant lesion severity and progression - Google Patents

Methods related to bronchial premalignant lesion severity and progression Download PDF

Info

Publication number
US11579140B2
US11579140B2 US16/545,032 US201916545032A US11579140B2 US 11579140 B2 US11579140 B2 US 11579140B2 US 201916545032 A US201916545032 A US 201916545032A US 11579140 B2 US11579140 B2 US 11579140B2
Authority
US
United States
Prior art keywords
module
gene
expression
subject
level
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
US16/545,032
Other languages
English (en)
Other versions
US20200057053A1 (en
Inventor
Jennifer E. Beane-Ebel
Avrum E. Spira
Marc Lenburg
Mary E. Reid
Sarah Mazzilli
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Boston University
Health Research Inc
Original Assignee
Boston University
Health Research Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Boston University, Health Research Inc filed Critical Boston University
Priority to US16/545,032 priority Critical patent/US11579140B2/en
Assigned to TRUSTEES OF BOSTON UNIVERSITY reassignment TRUSTEES OF BOSTON UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SPIRA, AVRUM E., LENBURG, Marc, MAZZILLI, SARAH, BEANE-EBEL, JENNIFER E.
Assigned to HEALTH RESEARCH, INC. reassignment HEALTH RESEARCH, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: REID, MARY E.
Publication of US20200057053A1 publication Critical patent/US20200057053A1/en
Priority to US18/093,066 priority patent/US20230146253A1/en
Application granted granted Critical
Publication of US11579140B2 publication Critical patent/US11579140B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B25/00ICT specially adapted for hybridisation; ICT specially adapted for gene or protein expression
    • G16B25/10Gene or protein expression profiling; Expression-ratio estimation or normalisation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/502Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects
    • G01N33/5023Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects on expression patterns
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5091Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing the pathological state of an organism
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57423Specifically defined cancers of lung
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B20/00ICT specially adapted for functional genomics or proteomics, e.g. genotype-phenotype associations
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B40/00ICT specially adapted for biostatistics; ICT specially adapted for bioinformatics-related machine learning or data mining, e.g. knowledge discovery or pattern finding
    • G16B40/20Supervised data analysis
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B40/00ICT specially adapted for biostatistics; ICT specially adapted for bioinformatics-related machine learning or data mining, e.g. knowledge discovery or pattern finding
    • G16B40/30Unsupervised data analysis
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H20/00ICT specially adapted for therapies or health-improving plans, e.g. for handling prescriptions, for steering therapy or for monitoring patient compliance
    • G16H20/10ICT specially adapted for therapies or health-improving plans, e.g. for handling prescriptions, for steering therapy or for monitoring patient compliance relating to drugs or medications, e.g. for ensuring correct administration to patients
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H50/00ICT specially adapted for medical diagnosis, medical simulation or medical data mining; ICT specially adapted for detecting, monitoring or modelling epidemics or pandemics
    • G16H50/20ICT specially adapted for medical diagnosis, medical simulation or medical data mining; ICT specially adapted for detecting, monitoring or modelling epidemics or pandemics for computer-aided diagnosis, e.g. based on medical expert systems
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H50/00ICT specially adapted for medical diagnosis, medical simulation or medical data mining; ICT specially adapted for detecting, monitoring or modelling epidemics or pandemics
    • G16H50/30ICT specially adapted for medical diagnosis, medical simulation or medical data mining; ICT specially adapted for detecting, monitoring or modelling epidemics or pandemics for calculating health indices; for individual health risk assessment
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/112Disease subtyping, staging or classification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Definitions

  • the technology described herein relates to treatment, diagnosis, and monitoring of treatment for bronchial premalignant lesions.
  • Lung squamous cell cancer develops from non-cancerous lesions in the airway known as bronchial premalignant lesions.
  • bronchial premalignant lesions The presence of persistent or progressive dysplastic bronchial premalignant lesions is a marker of increased lung cancer risk both at the lesion site (where they are the presumed precursors of squamous cell lung cancer) and elsewhere in the lung. Not all bronchial premalignant lesions progress to invasive cancer, and those that do, progress at variable rates with variable outcomes. At present, there are no tools available in the clinic to identify which lesions will progress to cancer and which will not. Additionally, the current technology for detecting bronchial premalignant lesions is via autofluorescence and white-light bronchoscopy.
  • a bronchoscopy procedure is invasive and is only moderately sensitive and specific at detecting small bronchial premalignant lesions as it requires visualization of the lesions. Finally, to date, the only treatment for bronchial premalignant lesions is to remove the lesions via surgery or bronchoscopy.
  • the inventors have now developed: 1) tests for the presence of bronchial premalignant lesions (some of which do not require bronchoscopy and use the surprising finding that normal tissues elsewhere in the airway exhibit biomarkers indicating the presence of bronchial premalignant lesions in the subject), 2) methods for determining if the bronchial premalignant lesions is likely to progress to cancer, 3) new therapies for bronchial premalignant lesions which target the underlying molecular changes which characterize the bronchial premalignant lesions.
  • one aspect is a method of treating bronchial premalignant lesions, the method comprising: administering at least one of: (i) both a bronchoscopy-based procedure to survey the central airway and a chest CT scan; (ii) at least every 6 months, one of a bronchoscopy-based procedure to survey the central airway and a chest CT scan; and/or (iii) at least one anti-proliferative drug; to a subject determined to have at least one of: (a) an increased level of expression of at least one module 5 gene as compared to a non-proliferative lesion reference level; and (b) a decreased level of expression of at least one module 6 gene as compared to a non-proliferative lesion reference level.
  • the at least one module 5 gene is selected from the group consisting of: RACGAP1 and TPX2; and the at least one module 6 gene is selected from the group consisting of: NEK11 and IFT88.
  • the subject is further determined to have an increased level of expression of at least one module 7 or module 4 gene.
  • the at least one module 7 or module 4 gene is selected from the group consisting of: COX6A1; COX7A2; RPL26; and RPL23.
  • the level of expression of each of the genes of Table 15 is determined.
  • the at least one anti-proliferative drug is selected from the group consisting of: Acetylcholine receptor antagonist; Acetylcholinesterase inhibitors; Adenosine receptor antagonists; Adrenergic receptor antagonists; AKT inhibitors; Angiotensin receptor antagonists; Apoptosis stimulants; Aurora kinase inhibitors; CDK inhibitors; Cyclooxygenase inhibitors; Cytokine production inhibitors; Dehydrogenase inhibitors; DNA protein kinase inhibitors; focal adhesion inhibitors; Dopamine receptor antagonist; EGFR inhibitors; ERK1 and ERK2 phosphorylation inhibitors; Estrogen receptor agonists; EZH2 inhibitors; FLT3 inhibitors; Glucocorticoid receptor agonists; Glutamate receptor antagonists; HDAC inhibitors; His
  • the anti-proliferative drug is administered as an inhaled formulation or topical formulation.
  • the anti-proliferative drug is administered during a bronchoscopy-based procedure.
  • the anti-proliferative drug is administered systemically.
  • the anti-proliferative drug is administered during a bronchoscopy-based procedure and systemically.
  • Another aspect provided herein relates to a method of treating bronchial premalignant lesions, the method comprising: administering at least one of: (i) both a bronchoscopy-based procedure to survey the central airway and a chest CT scan; (ii) at least every 6 months, one of a bronchoscopy-based procedure to survey the central airway and a chest CT scan; and/or (iii) at least one anti-proliferative drug; to a subject determined to have at least one of: (a) an increased level of expression of at least one module 5 gene as compared to a non-proliferative lesion reference level; and (b) a decreased level of expression of at least one module 6 gene as compared to a non-proliferative lesion reference level, wherein the subject is further determined to have a decreased level of expression of at least one module 9 gene as compared to a non-proliferative lesion reference level and/or an increased level of expression of at least one module 10 gene as compared to a non
  • the subject determined to have a decreased level of expression of at least one module 9 gene and/or an increased level of expression of at least one module 10 gene is administered at least one of:
  • a method of treating bronchial premalignant lesions comprising: administering at least one of: (i) both a bronchoscopy-based procedure to survey the central airway wherein the lesions are biopsied to remove abnormal tissue and a chest CT scan; (ii) at least every 6 months, one of a bronchoscopy-based procedure to survey the central airway wherein the lesions are biopsied to remove abnormal tissue and a chest CT scan; and/or (iii) at least one immune stimulating drug; to a subject determined to have a decreased level of expression of at least one module 9 gene as compared to a non-proliferative lesion reference level and/or an increased level of expression of at least one module 10 gene as compared to a non-proliferative lesion reference level.
  • the module 9 gene is selected from the group consisting of: EPSTI1; UBE2L6; B2M and TAP1.
  • the at least one gene module 9 gene is selected from Table 16.
  • the module 10 gene is selected from the group consisting of: CACNB3 and MAPK10.
  • the at least one immune stimulating drug is selected from the group consisting of: immune-checkpoint inhibitors (e.g. inhibitors against, PD-1, PD-L1, CTLA4, and LAG3); drugs that stimulate interferon signaling (e.g.
  • DNA synthesis inhibitors DNA synthesis inhibitors; IMDH inhibitors; CDK inhibitors; ribonucleotide reductase inhibitors; dihydrofolate reductase inhibitors; topoisomerase inhibitors; FLT3 inhibitors; IGF-1 inhibitors; MEK inhibitors; aurora kinase inhibitors; PKC inhibitors; RAF inhibitors; PDFGR/KIT inhibitors; VEGFR inhibitors; SRC inhibitors; retinoid receptor agonists; HDAC inhibitors; DNA methyltransferase inhibitors; and EZH2 inhibitors.
  • Another aspect provided herein relates to a method of treating bronchial premalignant lesions, the method comprising: administering at least one of: (i) both a bronchoscopy-based procedure to survey the central airway and a chest CT scan; (ii) at least every 6 months, one of a bronchoscopy-based procedure to survey the central airway and a chest CT scan; and/or (iii) at least one anti-inflammatory drug; to a subject determined to have at least one of: (a) an increased level of expression of at least one module 2 gene as compared to a non-inflammatory reference level; and (b) a decreased level of expression of at least one module 6 gene as compared to a non-inflammatory reference level.
  • the at least one module 2 gene is selected from the group consisting of: MSANTD2, CCNL2, and LUC7L; and the at least one module 6 gene is selected from the group consisting of: NEK11 and IFT88.
  • the subject is further determined to have an increased level of expression of at least one module 7 gene, module 1 gene, or module 8 gene and/or decreased level of expression of at least one module 4 gene or one module 5 gene.
  • the at least one module 7 gene is selected from the group consisting of: RPL26 and RPL23.
  • the at least one module 1 gene is selected from the group consisting of: KIRREL; PHLDB1; and MARVELD1.
  • the at least one module 8 gene is selected from the group consisting of: DOC2; CD53; and LAPTM.
  • the at least one module 4 gene is selected from the group consisting of: COX6A1 and COX7A2
  • the at least one module 5 gene is selected from the group consisting of: RACGAP1 and TPX2
  • the level of expression of each of the genes of Table 15 is determined.
  • the at least one anti-inflammatory drug is selected from the group consisting of: Acetylcholine receptor antagonists; Acetylcholinesterase inhibitors; Adenosine receptor antagonists; Adrenergic receptor antagonists; Angiotensin receptor antagonists; Anti-IL1B antibodies; Apoptosis stimulants; Aurora kinase inhibitors; CDK inhibitors; Cyclooxygenase inhibitors; Cytokine production inhibitors; Dehydrogenase inhibitors; Dopamine receptor antagonists; EGFR inhibitors; ERK1 and ERK2 phosphorylation inhibitors; Estrogen receptor agonists; FLT3 inhibitors; Glucocorticoid receptor agonists; Glutamate receptor antagonists; HDAC inhibitors; Histamine receptor antagonists; Histone lysine methyltransferase inhibitors; HSP inhibitors; IKK inhibitors; Ion channel antagonists; KIT inhibitors; Leucine rich repeat kinas
  • the anti-inflammatory drug is administered during a bronchoscopy-based procedure.
  • the anti-inflammatory drug is administered systemically.
  • the anti-inflammatory drug is administered during a bronchoscopy-based procedure and systemically.
  • the at least one gene is selected from Table 14.
  • the level of expression of each of the genes of Table 14 is determined.
  • lung cancer is lung squamous cell carcinoma.
  • the level of expression is the level of expression in an endobronchial biopsy, endobronchial brushing sample, large airway biopsy, large airway brushing sample, nasal epithelial cells, sputum, or blood obtained from the subject.
  • the level of expression is the level of expression in a bronchial brushing obtained from the right or left mainstem bronchus.
  • the biopsy or brushing sample comprises morphologically-normal tissues or cells.
  • the biopsy or brushing sample consists of morphologically-normal tissues or cells.
  • the level of expression is the level of expression in a sample comprising bronchial premalignant lesion cells.
  • the level of expression is the level of expression in a sample comprising morphologically-normal cells.
  • the subject is a smoker or former smoker.
  • FIGS. 1 A- 1 E demonstrate that endobronchial biopsies divide into four distinct molecular subtypes that correlate with clinical and molecular phenotypes.
  • the heatmap shows semi-supervised hierarchal clustering of z-score normalized gene expression across the 3,936 genes and 190 DC biopsies.
  • FIG. 1 B Bubbleplots showing significant associations (p ⁇ 0.01 by Fisher's Exact Test) between the molecular subtypes and smoking status, biopsy histological grade, and the predicted LUSC tumor molecular subtypes.
  • the columns represent the 4 molecular subtypes (Proliferative, Inflammatory, Secretory, and Normal-like) and the diameter of the circle is proportional to the number of samples within each subtype that have the row phenotype.
  • FIG. 1 E Immunofluorescent staining demonstrating the increased MKI67 and KRT5 staining and reduced TUB1A1 staining in the Proliferative subtype in concordance with the expression of the corresponding marker genes.
  • the representative samples shown for the Proliferative and Inflammatory subtypes have dysplasia histology while the samples shown for the Secretory and Normal-like subtypes have normal histology (Magnification 200 ⁇ ).
  • FIGS. 2 A- 2 D demonstrate that phenotypic associations with the molecular subtypes are confirmed in an independent sample set.
  • FIG. 2 A The 190 DC biopsies and the 3,936 genes were used to build a 22-gene nearest centroid molecular subtype classifier. Semi-supervised hierarchal clustering of z-score normalized gene expression across the 22 classifier genes and 190 DC biopsies training samples.
  • FIG. 2 B The 22-gene nearest centroid molecular subtype classifier was used to predict the molecular subtypes of the 105 VC biopsies. Semi-supervised hierarchal clustering of z-score normalized gene expression across 22 genes and 105 VC is plotted.
  • FIG. 2 C Bubbleplots showing significant associations (p ⁇ 0.01 by Fisher's Exact Test) between the VC molecular subtypes and smoking status, biopsy histological grade, and the predicted LUSC tumor molecular subtypes.
  • the columns represent the 4 molecular subtypes (Proliferative, Inflammatory, Secretory, and Normal) and the radius of the circle is proportional to the number of samples within each subtype that have the row phenotype.
  • FIGS. 3 A- 3 C demonstrate the performance of the molecular subtype classifier in the large airway brushes from normal appearing epithelium sampled at the same time as the endobronchial biopsies.
  • FIG. 3 A The DC (left) and VC (right) cohorts, showing the number of brushes (y-axis) predicted to be positive for the Proliferative subtype that have at least one biopsy (y-axis) with a classification of the Proliferative subtype at the time the brush was sampled.
  • FIG. 3 B Boxplots of PC1 for Modules 4, 5, 6, and 7 (y-axis) across the four molecular subtypes for each cohort (x-axis).
  • FIG. 3 C Boxplots of PC1 for Modules 4, 5, 6, and 7 (y-axis) across the four molecular subtypes for each cohort (x-axis). The asterisk indicates significant differences between the Proliferative subtype versus all other samples (FDR ⁇ 0.05).
  • FIGS. 4 A- 4 H demonstrate that the module enriched for interferon signaling and antigen processing is associated with biopsy progression/persistence and a depletion of innate and adaptive immune cells in the Proliferative subtype.
  • FIGS. 4 B and 4 G Boxplot of the percentages of CD68 and CD163, CD68, CD163, CD4, and CD8 positively stained cells between progressive/persistent and regressive biopsies (p ⁇ 0.001 for all comparisons).
  • the x-axis labels indicate the number of regions (R) enumerated across (P) subjects for each stain and outcome group depicted in the boxplot. Biopsies were included in the analysis if their clinical outcome was concordant with the Module 9 score.
  • FIG. 4 C Top: Z-score normalized gene expression across the 112 genes in Module 9 and the DC biopsies (left) and the VC biopsies (right). Each heatmap is supervised according to the Module 9 GSVA scores.
  • Top bars indicate the histological grade of the biopsies and their progression status.
  • FIG. 4 D Representative histology where the dashed line denoted the separate of epithelium and stromal compartment Top panels: A progressive severe dysplasia has reduced presence of immune cells demonstrated by the marked reduction in expression of M2 macrophages (CD68/163 staining, double positive cells indicated by the arrows) and CD8 T cells.
  • FIGS. 4 E and 4 H Boxplots of the percentages of CD68 and CD163, CD68, CD163, CD4, and CD8 positively stained cells between progressive/persistent and regressive biopsies (p ⁇ 0.001 for all comparisons).
  • the x-axis labels indicate the number of regions (R) enumerated across (P) subjects for each stain and outcome group depicted in the boxplot. Biopsies were included in the analysis if their clinical outcome was concordant with the Module 9 score.
  • FIG. 5 depicts Batch Information and Alignment Statistics on Samples in both the Discovery and Validation cohorts. Statistical tests between the Discovery and Validation cohorts were performed using Fisher's Exact Test for categorical variables and Student's T-Test for continuous variable. Percentages are reported for categorical variables and mean and standard deviations are reported for continuous variables.
  • FIG. 6 depicts a summary of Gene Modules. The module number, number of genes in the module, biological pathways and select genes associated with the module, and an FDR value for the difference in GSVA scores for the module between the molecular subtypes are reported.
  • FIG. 7 depicts a List of Samples used for Immunofluorescence Studies.
  • FIG. 8 depicts the distribution of Molecular Subtypes by Subject.
  • the columns represent the 4 molecular subtypes (Proliferative, Inflammatory, Secretory, and Normal-like) and the radius of the circle is proportional to the number of samples within each subtype.
  • FIGS. 10 A- 10 H depict boxplots of Select Genes and Cell Type Deconvolution Results across the Discovery and Validation Cohorts by Molecular Subtype.
  • FIGS. 10 A- 10 D Discovery cohort biopsies.
  • FIGS. 10 E- 10 H Validation cohort biopsies.
  • FIG. 10 A and ( FIG. 10 E ) show boxplots of gene expression levels of LUSC driver genes identified by TCGA across the molecular subtypes.
  • FIG. 10 B and ( FIG. 10 F ) show boxplots of gene expression levels of cell type marker genes across the molecular subtypes.
  • FIG. 10 C and ( FIG.
  • FIG. 10 G show boxplots of GSVA scores calculated using Dvorak et al. gene sets across the molecular subtypes.
  • FIG. 10 D shows boxplots of ESTIMATE algorithm scores across the molecular subtypes.
  • the ESTIMATE algorithm estimates the stromal (StromalScore), immune (Immune Score), and epithelial (ESTIMATEScore) cell fractions in each sample. High immune and stromal scores indicate a high fraction of stromal and immune cells while low epithelial scores indicate a high fraction of epithelial cells.
  • FIG. 11 depicts a heatmap of the 22-gene Molecular Subtype Classifier in the Discovery and Validation Cohort Biopsies.
  • the rows of the heatmap show the gene module membership.
  • the first column color bar shows molecular subtype membership in the DC and the 22-gene predict subtype membership in the VC.
  • the second column color bar depicts correct and incorrect predictions in the DC using the 22-gene classifier and molecular subtypes derived by performing consensus clustering across the VC.
  • FIG. 12 depicts graphs of gene module behavior across the Molecular Subtypes in the Discovery and Validation Cohort Biopsies. The mean of the first principal component calculated across module genes is plotted for each molecular subtype.
  • FIG. 13 depicts the concordance between Module 9 and two Cell Type Deconvolution Analyses.
  • Top Hierarchal clustering of z-score normalized gene expression across the 112 genes in module 9 and the DC biopsies (left) and the VC biopsies (right). Each heatmap is supervised according to the module 9 GSVA scores. Top bars indicate the histological grade of the biopsies and their progression status.
  • FIG. 14 depicts a tracheobronchial map of the locations of the sites sampled by endobronchial biopsy.
  • FIG. 15 depicts the distribution of subject among the discovery cohort endobronchial biopsies across the four molecular subtypes.
  • the heatmap shows semi-supervised hierarchal clustering of z-score normalized gene expression across the 3,936 genes and 190 DC biopsies.
  • the mean module GSVA score is plotted for each subtype.
  • FIG. 16 depicts the molecular subtype distribution for each subject across bronchoscopy procedures.
  • the barplot shows for each subject and each bronchoscopy procedure the number of biopsies sampled and their corresponding molecular subtype.
  • the y-axis indicates the subject number and whether or not that subject had a prior history of either lung squamous cell carcinoma (LUSC) or another type of lung cancer (Other).
  • the discovery cohort includes subjects 1 through 32 and the validation cohort includes subjects 33 through 52.
  • premalignant lesions in the airway of a subject can be characterized as being one of five: types: normal-like, secretory, inflammatory, progressive proliferative, and persistent proliferative. Identifying the premalignant lesion as one of these types permits more effective treatment of the subject, as different types of lesions will be responsive to different treatments and require different treatment and monitoring regimes. Accordingly, provided herein are methods of treatment relating to the treatment of bronchial premalignant lesions in a subject. Such methods can comprise assays, tests, and/or identification of the lesion type and administration of therapeutic regimens appropriate for that lesion type.
  • premalignant lesion refers to an epithelial lesion or dysplasia which is a precursor or can be a precursor to cancer.
  • the basement membrane is intact with no possibility of metastatic spread, as opposed to cancer.
  • a bronchial premalignant lesion is a premalignant lesion present in the bronchial epithelium of a subject. Bronchial premalignant lesions are typically small and can be difficult to visualize using conventional white light bronchoscopy.
  • the bronchial premalignant lesions can exhibit one of five phenotypes described herein, namely progressive proliferative, persistent proliferative, secretory, inflammatory, and normal-like.
  • the subtype names reference the key differences in molecular pathway activity which differentiate the subtypes from each other.
  • the different phenotypes of lesion can be distinguished from each other and from normal tissue by use of the gene expression patterns described herein.
  • the gene expression patterns identified herein relate to 10 modules of genes, where each module is a group of genes with similar expression patterns across the different bronchial premalignant lesion subtypes. The identity of each of the modules, e.g. the genes that comprise each module, are provided in Table 13 herein.
  • proliferative lesions are distinguished by having increased module 4, 5, and 7 expression and decreased module 6 expression.
  • Progressive proliferative lesions can be distinguished from persistent proliferative lesions in that they have decreased module 9 expression and/or increased module 10 expression.
  • Secretory lesions are distinguished by an increase in module 6 expression and a decrease in module 1 expression and optionally, an increase in module 8 expression and a decrease in modules 2, 5, and 7 expression.
  • Normal-like subtype is distinguished by an increase in module 6 expression and a decrease in module 9 expression and optionally, an increase in module 1 expression and a decrease in module 8 expression.
  • Standard treatment for subjects at risk of lung cancer, or who have been identified to have bronchial premalignant lesions is annual screening for lung cancer (e.g. a bronchoscopy and/or chest CT scan).
  • lung cancer e.g. a bronchoscopy and/or chest CT scan.
  • a method of treating bronchial premalignant lesions comprising administering at least one of: i) both a bronchoscopy-based procedure to survey the central airway and a chest CT scan; ii) at least every 6 months, at least one of a bronchoscopy-based procedure to survey the central airway and a chest CT scan; and/or iii) at least one anti-proliferative drug to a subject determined to have at least one of a) an increased level of expression of at least one module 5 gene as compared to a reference level; and b) a decreased level of expression of at least one module 6 gene as compared to a reference level.
  • a method of treating bronchial premalignant lesions comprising determining a subject as to have at least one of a) an increased level of expression of at least one module 5 gene as compared to a reference level; and b) a decreased level of expression of at least one module 6 gene as compared to a reference level and administering at least one of: i) both a bronchoscopy-based procedure to survey the central airway and a chest CT scan; ii) at least about every 6 months (e.g., at least every 1, 2, 3, 4, 5, or 6 months), at least one of a bronchoscopy-based procedure to survey the central airway and a chest CT scan; and/or iii) at least one anti-proliferative drug to the subject.
  • the reference level is a non-proliferative reference level.
  • the subject is not administered an anti-proliferative drug and is administered a bronchoscopy-based procedure to survey the central airway and/or a chest CT scan no more frequently than every 6 months (e.g., no more frequently than every 6, 7, 8, 9, 10, 11, or 12 months).
  • the subject is not administered an anti-proliferative drug and is administered a bronchoscopy-based procedure to survey the central airway and/or a chest CT scan no more frequently than every 6 months (e.g., no more frequently than every 6, 7, 8, 9, 10, 11, or 12 months).
  • Module 5 and 6 gene expression, in a bronchial brushing sample is sufficient to identify a subject having a proliferative subtype lesion. This avoids the need to visualize and/or sample the actual lesion. Accordingly, in one aspect of any of the embodiments, provided herein is a method of treating bronchial premalignant lesions, the method comprising administering at least one of: i) both a bronchoscopy-based procedure to survey the central airway and a chest CT scan; ii) at least every 6 months, at least one of a bronchoscopy-based procedure to survey the central airway and a chest CT scan; and/or iii) at least one anti-proliferative drug to a subject determined to have, in a bronchial brushing sample, a) an increased level of expression of at least one module 5 gene as compared to a reference level; and b) a decreased level of expression of at least one module 6 gene as compared to a reference level.
  • a method of treating bronchial premalignant lesions comprising determining a subject to have, in a bronchial brushing sample obtained from the subject, a) an increased level of expression of at least one module 5 gene as compared to a reference level; and b) a decreased level of expression of at least one module 6 gene as compared to a reference level and administering at least one of: i) both a bronchoscopy-based procedure to survey the central airway and a chest CT scan; ii) at least every 6 months, at least one of a bronchoscopy-based procedure to survey the central airway and a chest CT scan; and/or iii) at least one anti-proliferative drug to the subject.
  • the reference level is a non-proliferative reference level.
  • the bronchial brushing is taken from a morphologically-normal location in the right or left mainstem bronchus. In some embodiments of any of the aspects, the bronchial brushing is taken from a visually-normal location in the right or left mainstem bronchus.
  • Module 5 and 6 genes are provided in Table 13.
  • the at least one module 5 gene and/or module 6 gene can be any one or more of the module 5 and 6 genes listed in Table 13.
  • the level of expression of at least one module 5 gene or at least one module 6 gene is determined. In some embodiments of any of the aspects, the level of expression of two or more module 5 genes or two or more module 6 genes is determined. In some embodiments of any of the aspects, the level of expression of each module 5 gene or each module 6 gene of Table 13 is determined.
  • the level of expression of at least one module 5 gene and at least one module 6 gene is determined. In some embodiments of any of the aspects, the level of expression of two or more module 5 genes and two or more module 6 genes is determined. In some embodiments of any of the aspects, the level of expression of each module 5 gene and each module 6 gene of Table 13 is determined.
  • the at least one module 5 gene comprises or is RACGAP1 or TPX2. In some embodiments of any of the aspects, the at least one module 5 gene comprises or is RACGAP1 and TPX2. In some embodiments of any of the aspects, the at least one module 6 gene comprises or is NEK11 or IFT88. In some embodiments of any of the aspects, the at least one module 6 gene comprises or is NEK11 and IFT88.
  • the proliferative subtype is further distinguished by increased expression of module 7 and/or 4. Accordingly, in some embodiments of any of the aspects, the subject is further determined to have an increased level of expression of at least one module 7 or module 4 gene as compared to a reference level. In some embodiments of any of the aspects, the subject is further determined to have an increased level of expression of at least one module 7 and at least one module 4 gene as compared to a reference level. In some embodiments of any of the aspects, the reference level is a non-proliferative reference level.
  • Module 4 and 7 genes are provided in Table 13.
  • the at least one module 4 gene and/or module 7 gene can be any one or more of the module 4 and 7 genes listed in Table 13.
  • the level of expression of at least one module 4 gene or at least one module 7 gene is determined. In some embodiments of any of the aspects, the level of expression of two or more module 4 genes or two or more module 7 genes is determined. In some embodiments of any of the aspects, the level of expression of each module 4 gene or each module 7 gene of Table 13 is determined.
  • the level of expression of at least one module 4 gene and at least one module 7 gene is determined. In some embodiments of any of the aspects, the level of expression of two or more module 4 genes and two or more module 7 genes is determined. In some embodiments of any of the aspects, the level of expression of each module 4 gene and each module 7 gene of Table 13 is determined.
  • the at least one module 4 gene comprises or is COX6A1 or COX7A2. In some embodiments of any of the aspects, the at least one module 4 gene comprises or is COX6A1 and COX7A2. In some embodiments of any of the aspects, the at least one module 7 gene comprises or is RPL26 or RPL23. In some embodiments of any of the aspects, the at least one module 7 gene comprises or is RPL26 and RPL23.
  • a method of treating bronchial premalignant lesions comprising administering at least one of: i) both a bronchoscopy-based procedure to survey the central airway and a chest CT scan; ii) at least every 6 months, at least one of a bronchoscopy-based procedure to survey the central airway and a chest CT scan; iii) at least one immune stimulating drug and/or iv) at least one immune stimulating drug and at least one anti-proliferative drug to a subject determined to have a decreased level of expression of at least one module 9 gene as compared to a reference level and/or an increased level of expression of at least one module 10 gene as compared to a reference level.
  • a method of treating bronchial premalignant lesions comprising a) determining a subject as to have a decreased level of expression of at least one module 9 gene as compared to a reference level and/or an increased level of expression of at least one module 10 gene as compared to a reference level and b) administering at least one of: i) both a bronchoscopy-based procedure to survey the central airway and a chest CT scan; ii) at least every 6 months, at least one of a bronchoscopy-based procedure to survey the central airway and a chest CT scan; iii) at least one immune stimulating drug and/or iv) at least one immune stimulating drug and at least one anti-proliferative drug to the subject.
  • the reference level is a non-proliferative reference level.
  • the bronchoscopy-based procedure further comprises biopsy of the lesions to remove
  • the subject if the subject is determined not to have a decreased level of expression of at least one module 9 gene as compared to a reference level and/or not to have an increased level of expression of at least one module 10 gene as compared to a reference level, the subject i) is not administered an immune stimulating drug, ii) is not administered both an immune stimulating drug and an anti-proliferative drug, iii) is administered a bronchoscopy-based procedure and/or a chest CT scan no more frequently than every 6 months (e.g., no more frequently than every 6, 7, 8, 9, 10, 11, or 12 months), and/or iv) is not administered a bronchoscopy-based procedure to biopsy lesions to remove abnormal tissue.
  • Module 9 genes are provided in Table 13.
  • the at least one module 9 gene can be any one or more of the module 9 genes listed in Table 13.
  • Module 9 genes are provided in Table 16.
  • the at least one module 9 gene can be any one or more of the module 9 genes listed in Table 16.
  • the level of expression of two or more module 9 gene is determined. In some embodiments of any of the aspects, the level of expression of each module 9 gene of Table 13 is determined. In some embodiments of any of the aspects, the level of expression of each module 9 gene of Table 16 is determined.
  • the at least one module 9 gene comprises or is EPSTI1; UBE2L6; B2M and/or TAP1. In some embodiments of any of the aspects, the at least one module 9 gene comprises or is EPSTI1; UBE2L6; B2M; and TAP1. In some embodiments of any of the aspects, the at least one module 9 gene comprises or is a pairwise combination of any of:
  • Module 10 genes are provided in Table 13.
  • the at least one module 10 gene can be any one or more of the module 9 genes listed in Table 13. In some embodiments of any of the aspects, the level of expression of both module 10 genes is determined. In some embodiments of any of the aspects, the at least one module 10 gene comprises or is CACNB3 or MAPK10. In some embodiments of any of the aspects, the at least one module 10 gene comprises or is CACNB3 and MAPK10.
  • a method of treating bronchial premalignant lesions comprising administering at least one of: i) both a bronchoscopy-based procedure to survey the central airway and a chest CT scan; ii) at least every 6 months, at least one of a bronchoscopy-based procedure to survey the central airway and a chest CT scan; and/or iii) at least one anti-inflammatory drug to a subject determined to have at least one of a) an increased level of expression of at least one module 2 gene as compared to a reference level; and b) a decreased level of expression of at least one module 6 gene as compared to a reference level.
  • a method of treating bronchial premalignant lesions comprising determining a subject as to have at least one of a) an increased level of expression of at least one module 2 gene as compared to a reference level; and b) a decreased level of expression of at least one module 6 gene as compared to a reference level and administering at least one of: i) both a bronchoscopy-based procedure to survey the central airway and a chest CT scan; ii) at least every 6 months, at least one of a bronchoscopy-based procedure to survey the central airway and a chest CT scan; and/or iii) at least one anti-inflammatory drug to the subject.
  • the reference level is a non-inflammatory reference level.
  • the subject is determined not to have at least one of a) an increased level of expression of at least one module 2 gene as compared to a reference level; and b) a decreased level of expression of at least one module 6 gene as compared to a reference level, the subject is not administered an anti-inflammatory drug and is administered a bronchoscopy-based procedure to survey the central airway and/or a chest CT scan no more frequently than every 6 months (e.g., no more frequently than every 6, 7, 8, 9, 10, 11, or 12 months).
  • the subject is not administered an anti-inflammatory drug and is administered a bronchoscopy-based procedure to survey the central airway and/or a chest CT scan no more frequently than every 6 months (e.g., no more frequently than every 6, 7, 8, 9, 10, 11, or 12 months).
  • Module 2 and 6 genes are provided in Table 13.
  • the at least one module 2 gene and/or module 6 gene can be any one or more of the module 5 and 6 genes listed in Table 13.
  • the level of expression of at least one module 2 gene or at least one module 6 gene is determined. In some embodiments of any of the aspects, the level of expression of two or more module 2 genes or two or more module 6 genes is determined. In some embodiments of any of the aspects, the level of expression of each module 2 gene or each module 6 gene of Table 13 is determined.
  • the level of expression of at least one module 2 gene and at least one module 6 gene is determined. In some embodiments of any of the aspects, the level of expression of two or more module 2 genes and two or more module 6 genes is determined. In some embodiments of any of the aspects, the level of expression of each module 2 gene and each module 6 gene of Table 13 is determined.
  • the at least one module 2 gene comprises or is MSANTD2, CCNL2, or LUC7L. In some embodiments of any of the aspects, the at least one module 2 gene comprises or is MSANTD2 and LUC7L. In some embodiments of any of the aspects, the at least one module 2 gene comprises or is MSANTD2 and CCNL2. In some embodiments of any of the aspects, the at least one module 2 gene comprises or is CCNL2 and LUC7L. In some embodiments of any of the aspects, the at least one module 2 gene comprises or is MSANTD2, CCNL2, and LUC7L. In some embodiments of any of the aspects, the at least one module 6 gene comprises or is NEK11 or IFT88. In some embodiments of any of the aspects, the at least one module 6 gene comprises or is NEK11 and IFT88.
  • the inflammatory subtype is further distinguished by increased expression of module 7, 1 and/or 8 and/or decreased expression of module 4 and/or 5. Accordingly, in some embodiments of any of the aspects, the subject is further determined to have at least one of: i) an increased level of expression of at least one module 7, module 1, and/or or module 8 gene, and ii) a decreased level of expression of at least one module 4 or module 5 gene as compared to a reference level. In some embodiments of any of the aspects, the subject is further determined to have at least one of: i) an increased level of expression of at least one module 7, module 1, and/or or module 8 gene, and ii) a decreased level of expression of at least one module 4 or module 5 gene as compared to a reference level. In some embodiments of any of the aspects, the reference level is a non-inflammatory reference level.
  • Module 7, 1, 8, 4 and 5 genes are provided in Table 13.
  • the at least one module 7, 1, 8, 4, and/or 5 gene can be any one or more of the module 7, 1, 8, 4, and/or 5 genes listed in Table 13.
  • the level of expression of each module 7, 1, 8, 4 and/or 5 gene of Table 13 is determined. In some embodiments of any of the aspects, the level of expression of each module 7, 1, 8, 4 and 5 gene of Table 13 is determined.
  • the at least one module 4 gene comprises or is COX6A1 or COX7A2. In some embodiments of any of the aspects, the at least one module 4 gene comprises or is COX6A1 and COX7A2. In some embodiments of any of the aspects, the at least one module 7 gene comprises or is RPL26 or RPL23. In some embodiments of any of the aspects, the at least one module 7 gene comprises or is RPL26 and RPL23. In some embodiments of any of the aspects, the at least one module 5 gene comprises or is RACGAP1 or TPX2. In some embodiments of any of the aspects, the at least one module 5 gene comprises or is RACGAP1 and TPX2.
  • the at least one module 1 gene comprises or is KIRREL; PHLDB1; or MARVELD1. In some embodiments of any of the aspects, the at least one module 1 gene comprises or is PHLDB1 and MARVELD1. In some embodiments of any of the aspects, the at least one module 1 gene comprises or is KIRREL and PHLDB1. In some embodiments of any of the aspects, the at least one module 1 gene comprises or is KIRREL and MARVELD1. In some embodiments of any of the aspects, the at least one module 1 gene comprises or is KIRREL; PHLDB1; and MARVELD1.
  • the at least one module 8 gene comprises or is DCO2; CD53; or LAPTM. In some embodiments of any of the aspects, the at least one module 8 gene comprises or is CD53 and LAPTM. In some embodiments of any of the aspects, the at least one module 8 gene comprises or is DCO2 and CD53. In some embodiments of any of the aspects, the at least one module 8 gene comprises or is DCO2 and LAPTM. In some embodiments of any of the aspects, the at least one module 8 gene comprises or is DCO2; CD53; and LAPTM.
  • the level of expression of each of the genes of Table 15 is determined. In some embodiments of any of the aspects, the level of expression of each of the genes of Table 15 in a bronchial brushing sample is determined.
  • the level of expression of each of the genes of Table 14 is determined. In some embodiments of any of the aspects, the level of expression of each of the genes of Table 14 in a bronchial brushing sample is determined.
  • the methods described herein can further comprise determining the level of expression of any of the following genes: SOX2, NFE2L2, PIK3CA (which are squamous cancer marker genes), KRT5, MUC5AC, TUB1A1, SCGB1A1, and FOXK1 (which are epithelial marker genes).
  • levels of gene expression can be modulated (e.g., increased or decreased) in subjects with premalignant lesions of different subtypes.
  • the method comprises administering a treatment described herein to a subject previously determined to have an expression level(s) as described herein.
  • described herein is a method of treating bronchial premalignant lesions in a subject in need thereof, the method comprising: a) first determining the level of expression of the at least one gene in a sample obtained from a subject; and b) then administering a treatment as described herein to the subject if the level of expression of modulated relative to a reference in the manner described herein.
  • described herein is a method of treating bronchial premalignant lesions in a subject in need thereof, the method comprising: a) determining if the subject has a modulation of a level of expression as described herein and b) instructing or directing that the subject be administered the appropriate treatment described herein for the particular modulation of expression which has been determined.
  • the step of determining if the subject has modulation of an expression level can comprise i) obtaining or having obtained a sample from the subject and ii) performing or having performed an assay on the sample obtained from the subject to determine/measure the level of expression in the subject. In some embodiments of any of the aspects, the step of determining if the subject has a modulation of a level of expression can comprise performing or having performed an assay on a sample obtained from the subject to determine/measure the level of expression in the subject. In some embodiments of any of the aspects, the step of determining if the subject has a modulation of a level of expression can comprise ordering or requesting an assay on a sample obtained from the subject to determine/measure the level of expression in the subject.
  • the step of determining if the subject has a modulation of a level of expression can comprise receiving the results of an assay on a sample obtained from the subject to determine/measure the level of expression in the subject. In some embodiments of any of the aspects, the step of determining if the subject has a modulation of a level of expression can comprise receiving a report, results, or other means of identifying the subject as a subject with a modulation of a level of expression.
  • the step of instructing or directing that the subject be administered a particular treatment can comprise providing a report of the assay results. In some embodiments of any of the aspects, the step of instructing or directing that the subject be administered a particular treatment can comprise providing a report of the assay results and/or treatment recommendations in view of the assay results.
  • measurement of the level of a target and/or detection of the level or presence of a target can comprise a transformation.
  • transforming or “transformation” refers to changing an object or a substance, e.g., biological sample, nucleic acid or protein, into another substance.
  • the transformation can be physical, biological or chemical. Exemplary physical transformation includes, but is not limited to, pre-treatment of a biological sample, e.g., from whole blood to blood serum by differential centrifugation.
  • a biological/chemical transformation can involve the action of at least one enzyme and/or a chemical reagent in a reaction.
  • a DNA sample can be digested into fragments by one or more restriction enzymes, or an exogenous molecule can be attached to a fragmented DNA sample with a ligase.
  • a DNA sample can undergo enzymatic replication, e.g., by polymerase chain reaction (PCR).
  • Transformation, measurement, and/or detection of a target molecule can comprise contacting a sample obtained from a subject with a reagent (e.g. a detection reagent) which is specific for the target, e.g., a target-specific reagent.
  • a reagent e.g. a detection reagent
  • the target-specific reagent is detectably labeled.
  • the target-specific reagent is capable of generating a detectable signal.
  • the target-specific reagent generates a detectable signal when the target molecule is present.
  • ELISA enzyme linked immunosorbent assay
  • western blot immunoprecipitation
  • immunofluorescence using detection reagents such as an antibody or protein binding agents.
  • detection reagents such as an antibody or protein binding agents.
  • a peptide can be detected in a subject by introducing into a subject a labeled anti-peptide antibody and other types of detection agent.
  • the antibody can be labeled with a detectable marker whose presence and location in the subject is detected by standard imaging techniques.
  • antibodies for the various targets described herein are commercially available and can be used for the purposes of the invention to measure protein expression levels.
  • amino acid sequences for the targets described herein are known and publically available at the NCBI website, one of skill in the art can raise their own antibodies against these polypeptides of interest for the purpose of the methods described herein.
  • amino acid sequences of the polypeptides described herein have been assigned NCBI and ENSBL accession numbers for different species such as human, mouse and rat.
  • sequences for any of the genes described herein can be readily retrieved from either database by one of ordinary skill in the art.
  • sequence of a gene, transcript, or polypeptide described herein is the sequence available in the NCBI or ENSMBL database as of the filing date of this application.
  • immunohistochemistry is the application of immunochemistry to tissue sections
  • ICC is the application of immunochemistry to cells or tissue imprints after they have undergone specific cytological preparations such as, for example, liquid-based preparations.
  • Immunochemistry is a family of techniques based on the use of an antibody, wherein the antibodies are used to specifically target molecules inside or on the surface of cells. The antibody typically contains a marker that will undergo a biochemical reaction, and thereby experience a change of color, upon encountering the targeted molecules.
  • signal amplification can be integrated into the particular protocol, wherein a secondary antibody, that includes the marker stain or marker signal, follows the application of a primary specific antibody.
  • the assay can be a Western blot analysis.
  • proteins can be separated by two-dimensional gel electrophoresis systems. Two-dimensional gel electrophoresis is well known in the art and typically involves iso-electric focusing along a first dimension followed by SDS-PAGE electrophoresis along a second dimension. These methods also require a considerable amount of cellular material.
  • the analysis of 2D SDS-PAGE gels can be performed by determining the intensity of protein spots on the gel, or can be performed using immune detection.
  • protein samples are analyzed by mass spectroscopy.
  • Immunological tests can be used with the methods and assays described herein and include, for example, competitive and non-competitive assay systems using techniques such as Western blots, radioimmunoassay (RIA), ELISA (enzyme linked immunosorbent assay), “sandwich” immunoassays, immunoprecipitation assays, immunodiffusion assays, agglutination assays, e.g. latex agglutination, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, e.g.
  • FIA fluorescence-linked immunoassay
  • CLIA chemiluminescence immunoassays
  • ELIA electrochemiluminescence immunoassay
  • CIA counting immunoassay
  • LFIA lateral flow tests or immunoassay
  • MIA magnetic immunoassay
  • protein A immunoassays Methods for performing such assays are known in the art, provided an appropriate antibody reagent is available.
  • the immunoassay can be a quantitative or a semi-quantitative immunoassay.
  • An immunoassay is a biochemical test that measures the concentration of a substance in a biological sample, typically a fluid sample such as blood or serum, using the interaction of an antibody or antibodies to its antigen.
  • the assay takes advantage of the highly specific binding of an antibody with its antigen.
  • specific binding of the target polypeptides with respective proteins or protein fragments, or an isolated peptide, or a fusion protein described herein occurs in the immunoassay to form a target protein/peptide complex.
  • the complex is then detected by a variety of methods known in the art.
  • An immunoassay also often involves the use of a detection antibody.
  • Enzyme-linked immunosorbent assay also called ELISA, enzyme immunoassay or EIA
  • ELISA enzyme immunoassay
  • EIA enzyme immunoassay
  • an ELISA involving at least one antibody with specificity for the particular desired antigen can also be performed.
  • a known amount of sample and/or antigen is immobilized on a solid support (usually a polystyrene micro titer plate). Immobilization can be either non-specific (e.g., by adsorption to the surface) or specific (e.g. where another antibody immobilized on the surface is used to capture antigen or a primary antibody). After the antigen is immobilized, the detection antibody is added, forming a complex with the antigen.
  • the detection antibody can be covalently linked to an enzyme, or can itself be detected by a secondary antibody which is linked to an enzyme through bio-conjugation.
  • the plate is typically washed with a mild detergent solution to remove any proteins or antibodies that are not specifically bound.
  • the plate is developed by adding an enzymatic substrate to produce a visible signal, which indicates the quantity of antigen in the sample.
  • Older ELISAs utilize chromogenic substrates, though newer assays employ fluorogenic substrates with much higher sensitivity.
  • a competitive ELISA is used.
  • Purified antibodies that are directed against a target polypeptide or fragment thereof are coated on the solid phase of multi-well plate, i.e., conjugated to a solid surface.
  • a second batch of purified antibodies that are not conjugated on any solid support is also needed.
  • These non-conjugated purified antibodies are labeled for detection purposes, for example, labeled with horseradish peroxidase to produce a detectable signal.
  • a sample e.g., a blood sample
  • a known amount of desired antigen e.g., a known volume or concentration of a sample comprising a target polypeptide
  • desired antigen e.g., a known volume or concentration of a sample comprising a target polypeptide
  • the mixture is then are added to coated wells to form competitive combination.
  • a complex of labeled antibody reagent-antigen will form. This complex is free in solution and can be washed away. Washing the wells will remove the complex.
  • TMB (3, 3′, 5, 5′-tetramethylbenzidene) color development substrate for localization of horseradish peroxidase-conjugated antibodies in the wells.
  • TMB 3, 3′, 5, 5′-tetramethylbenzidene
  • TMB 3, 3′, 5, 5′-tetramethylbenzidene
  • the levels of a polypeptide in a sample can be detected by a lateral flow immunoassay test (LFIA), also known as the immunochromatographic assay, or strip test.
  • LFIAs are a simple device intended to detect the presence (or absence) of antigen, e.g. a polypeptide, in a fluid sample.
  • LFIA tests are a form of immunoassay in which the test sample flows along a solid substrate via capillary action.
  • LFIAs are essentially immunoassays adapted to operate along a single axis to suit the test strip format or a dipstick format. Strip tests are extremely versatile and can be easily modified by one skilled in the art for detecting an enormous range of antigens from fluid samples such as urine, blood, water, and/or homogenized tissue samples etc.
  • Strip tests are also known as dip stick tests, the name bearing from the literal action of “dipping” the test strip into a fluid sample to be tested.
  • LFIA strip tests are easy to use, require minimum training and can easily be included as components of point-of-care test (POCT) diagnostics to be use on site in the field.
  • LFIA tests can be operated as either competitive or sandwich assays.
  • Sandwich LFIAs are similar to sandwich ELISA. The sample first encounters colored particles which are labeled with antibodies raised to the target antigen. The test line will also contain antibodies to the same target, although it may bind to a different epitope on the antigen. The test line will show as a colored band in positive samples.
  • the lateral flow immunoassay can be a double antibody sandwich assay, a competitive assay, a quantitative assay or variations thereof.
  • Competitive LFIAs are similar to competitive ELISA. The sample first encounters colored particles which are labeled with the target antigen or an analogue. The test line contains antibodies to the target/its analogue. Unlabelled antigen in the sample will block the binding sites on the antibodies preventing uptake of the colored particles. The test line will show as a colored band in negative samples.
  • lateral flow technology It is also possible to apply multiple capture zones to create a multiplex test.
  • a polypeptide or fragment thereof can be dissociated with detergents and heat, and separated on an SDS-PAGE gel before being transferred to a solid support, such as a nitrocellulose or PVDF membrane.
  • a solid support such as a nitrocellulose or PVDF membrane.
  • the membrane is incubated with an antibody reagent specific for the target polypeptide or a fragment thereof. The membrane is then washed to remove unbound proteins and proteins with non-specific binding.
  • Detectably labeled enzyme-linked secondary or detection antibodies can then be used to detect and assess the amount of polypeptide in the sample tested.
  • a dot blot immobilizes a protein sample on a defined region of a support, which is then probed with antibody and labelled secondary antibody as in Western blotting.
  • the intensity of the signal from the detectable label in either format corresponds to the amount of enzyme present, and therefore the amount of polypeptide.
  • Levels can be quantified, for example by densitometry.
  • the level of a target can be measured, by way of non-limiting example, by Western blot; immunoprecipitation; enzyme-linked immunosorbent assay (ELISA); radioimmunological assay (RIA); sandwich assay; fluorescence in situ hybridization (FISH); immunohistological staining; radioimmunometric assay; immunofluoresence assay; mass spectroscopy and/or immunoelectrophoresis assay.
  • Western blot immunoprecipitation
  • ELISA enzyme-linked immunosorbent assay
  • RIA radioimmunological assay
  • FISH fluorescence in situ hybridization
  • immunohistological staining radioimmunometric assay
  • immunofluoresence assay immunofluoresence assay
  • mass spectroscopy and/or immunoelectrophoresis assay can be measured, by way of non-limiting example, by Western blot; immunoprecipitation; enzyme-linked immunosorbent assay (ELISA); radioimmunological assay (RIA); sandwich assay; flu
  • the gene expression products as described herein can be instead determined by determining the level of messenger RNA (mRNA) expression of the genes described herein.
  • mRNA messenger RNA
  • Such molecules can be isolated, derived, or amplified from a biological sample, such as a blood sample.
  • Techniques for the detection of mRNA expression is known by persons skilled in the art, and can include but not limited to, PCR procedures, RT-PCR, quantitative RT-PCR Northern blot analysis, differential gene expression, RNAse protection assay, microarray based analysis, next-generation sequencing; hybridization methods, etc.
  • the PCR procedure describes a method of gene amplification which is comprised of (i) sequence-specific hybridization of primers to specific genes or sequences within a nucleic acid sample or library, (ii) subsequent amplification involving multiple rounds of annealing, elongation, and denaturation using a thermostable DNA polymerase, and (iii) screening the PCR products for a band of the correct size.
  • the primers used are oligonucleotides of sufficient length and appropriate sequence to provide initiation of polymerization, i.e. each primer is specifically designed to be complementary to a strand of the genomic locus to be amplified.
  • mRNA level of gene expression products described herein can be determined by reverse-transcription (RT) PCR and by quantitative RT-PCR (QRT-PCR) or real-time PCR methods.
  • RT reverse-transcription
  • QRT-PCR quantitative RT-PCR
  • real-time PCR methods Methods of RT-PCR and QRT-PCR are well known in the art.
  • the level of an mRNA can be measured by a quantitative sequencing technology, e.g. a quantitative next-generation sequence technology.
  • a quantitative sequencing technology e.g. a quantitative next-generation sequence technology.
  • Methods of sequencing a nucleic acid sequence are well known in the art. Briefly, a sample obtained from a subject can be contacted with one or more primers which specifically hybridize to a single-strand nucleic acid sequence flanking the target gene sequence and a complementary strand is synthesized.
  • an adaptor double or single-stranded
  • the sequence can be determined, e.g.
  • exemplary methods of sequencing include, but are not limited to, Sanger sequencing, dideoxy chain termination, high-throughput sequencing, next generation sequencing, 454 sequencing, SOLiD sequencing, polony sequencing, Illumina sequencing, Ion Torrent sequencing, sequencing by hybridization, nanopore sequencing, Helioscope sequencing, single molecule real time sequencing, RNAP sequencing, and the like. Methods and protocols for performing these sequencing methods are known in the art, see, e.g. “Next Generation Genome Sequencing” Ed.
  • nucleic acid sequences of the genes described herein have been assigned NCBI and ENSBL accession numbers for different species such as human, mouse and rat.
  • sequences for any of the genes described herein can be readily retrieved from either database by one of ordinary skill in the art.
  • sequence of a gene, transcript, or polypeptide described herein is the sequence available in the NCBI or ENSMBL database as of the filing date of this application. Accordingly, a skilled artisan can design an appropriate primer based on the known sequence for determining the mRNA level of the respective gene.
  • Nucleic acid and ribonucleic acid (RNA) molecules can be isolated from a particular biological sample using any of a number of procedures, which are well-known in the art, the particular isolation procedure chosen being appropriate for the particular biological sample.
  • freeze-thaw and alkaline lysis procedures can be useful for obtaining nucleic acid molecules from solid materials
  • heat and alkaline lysis procedures can be useful for obtaining nucleic acid molecules from urine
  • proteinase K extraction can be used to obtain nucleic acid from blood (Roiff, A et al. PCR: Clinical Diagnostics and Research, Springer (1994)).
  • one or more of the reagents can comprise a detectable label and/or comprise the ability to generate a detectable signal (e.g. by catalyzing reaction converting a compound to a detectable product).
  • Detectable labels can comprise, for example, a light-absorbing dye, a fluorescent dye, or a radioactive label. Detectable labels, methods of detecting them, and methods of incorporating them into reagents (e.g. antibodies and nucleic acid probes) are well known in the art.
  • detectable labels can include labels that can be detected by spectroscopic, photochemical, biochemical, immunochemical, electromagnetic, radiochemical, or chemical means, such as fluorescence, chemifluorescence, or chemiluminescence, or any other appropriate means.
  • the detectable labels used in the methods described herein can be primary labels (where the label comprises a moiety that is directly detectable or that produces a directly detectable moiety) or secondary labels (where the detectable label binds to another moiety to produce a detectable signal, e.g., as is common in immunological labeling using secondary and tertiary antibodies).
  • the detectable label can be linked by covalent or non-covalent means to the reagent.
  • a detectable label can be linked such as by directly labeling a molecule that achieves binding to the reagent via a ligand-receptor binding pair arrangement or other such specific recognition molecules.
  • Detectable labels can include, but are not limited to radioisotopes, bioluminescent compounds, chromophores, antibodies, chemiluminescent compounds, fluorescent compounds, metal chelates, and enzymes.
  • the detection reagent is label with a fluorescent compound.
  • a detectable label can be a fluorescent dye molecule, or fluorophore including, but not limited to fluorescein, phycoerythrin, phycocyanin, o-phthaldehyde, fluorescamine, Cy3TM, Cy5TM, allophycocyanine, Texas Red, peridenin chlorophyll, cyanine, tandem conjugates such as phycoerythrin-Cy5TM, green fluorescent protein, rhodamine, fluorescein isothiocyanate (FITC) and Oregon GreenTM, rhodamine and derivatives (e.g., Texas red and tetrarhodimine isothiocynate (TRITC)), biotin, phycoerythrin, AMCA, CyD
  • a detectable label can be a radiolabel including, but not limited to 3 H, 125 I, 35 S, 14 C, 32 P, and 33 P.
  • a detectable label can be an enzyme including, but not limited to horseradish peroxidase and alkaline phosphatase.
  • An enzymatic label can produce, for example, a chemiluminescent signal, a color signal, or a fluorescent signal.
  • Enzymes contemplated for use to detectably label an antibody reagent include, but are not limited to, malate dehydrogenase, staphylococcal nuclease, delta-V-steroid isomerase, yeast alcohol dehydrogenase, alpha-glycerophosphate dehydrogenase, triose phosphate isomerase, horseradish peroxidase, alkaline phosphatase, asparaginase, glucose oxidase, beta-galactosidase, ribonuclease, urease, catalase, glucose-VI-phosphate dehydrogenase, glucoamylase and acetylcholinesterase.
  • a detectable label is a chemiluminescent label, including, but not limited to lucigenin, luminol, luciferin, isoluminol, theromatic acridinium ester, imidazole, acridinium salt and oxalate ester.
  • a detectable label can be a spectral colorimetric label including, but not limited to colloidal gold or colored glass or plastic (e.g., polystyrene, polypropylene, and latex) beads.
  • detection reagents can also be labeled with a detectable tag, such as c-Myc, HA, VSV-G, HSV, FLAG, V5, HIS, or biotin.
  • a detectable tag such as c-Myc, HA, VSV-G, HSV, FLAG, V5, HIS, or biotin.
  • Other detection systems can also be used, for example, a biotin-streptavidin system.
  • the antibodies immunoreactive (i.e. specific for) with the biomarker of interest is biotinylated. Quantity of biotinylated antibody bound to the biomarker is determined using a streptavidin-peroxidase conjugate and a chromogenic substrate.
  • streptavidin peroxidase detection kits are commercially available, e.g.
  • a reagent can also be detectably labeled using fluorescence emitting metals such as 152 Eu, or others of the lanthanide series. These metals can be attached to the reagent using such metal chelating groups as diethylenetriaminepentaacetic acid (DTPA) or ethylene diaminetetraacetic acid (EDTA).
  • DTPA diethylenetriaminepentaacetic acid
  • EDTA ethylene diaminetetraacetic acid
  • the level of expression is the level in a sample obtained from a subject.
  • sample or “test sample” as used herein denotes a sample taken or isolated from a biological organism, e.g., a blood or tissue sample from a subject.
  • present invention encompasses several examples of a biological sample.
  • the biological sample is cells, or tissue, or peripheral blood, or bodily fluid.
  • Exemplary biological samples include, but are not limited to, a biopsy, a tumor sample, biofluid sample; blood; serum; plasma; urine; sperm; mucus; tissue biopsy; organ biopsy; synovial fluid; bile fluid; cerebrospinal fluid; mucosal secretion; effusion; sweat; saliva; and/or tissue sample etc.
  • the term also includes a mixture of the above-mentioned samples.
  • the term “test sample” also includes untreated or pretreated (or pre-processed) biological samples.
  • a test sample can comprise cells from a subject.
  • the sample obtained from a subject can be a biopsy sample.
  • the sample obtained from a subject can be a blood or serum sample.
  • the sample is an endobronchial biopsy, bronchial brushing sample, bronchial biopsy, endobronchial brushing sample, large airway biopsy, large airway brushing sample, nasal epithelial cells, sputum, and/or blood obtained from the subject.
  • the sample is a bronchial brushing obtained from the right or left mainstem bronchus.
  • the test sample can be obtained by removing a sample from a subject, but can also be accomplished by using a previously isolated sample (e.g. isolated at a prior timepoint and isolated by the same or another person).
  • the test sample can be an untreated test sample.
  • untreated test sample refers to a test sample that has not had any prior sample pre-treatment except for dilution and/or suspension in a solution.
  • Exemplary methods for treating a test sample include, but are not limited to, centrifugation, filtration, sonication, homogenization, heating, freezing and thawing, and combinations thereof.
  • the test sample can be a frozen test sample, e.g., a frozen tissue. The frozen sample can be thawed before employing methods, assays and systems described herein.
  • a frozen sample can be centrifuged before being subjected to methods, assays and systems described herein.
  • the test sample is a clarified test sample, for example, by centrifugation and collection of a supernatant comprising the clarified test sample.
  • a test sample can be a pre-processed test sample, for example, supernatant or filtrate resulting from a treatment selected from the group consisting of centrifugation, filtration, thawing, purification, and any combinations thereof.
  • the test sample can be treated with a chemical and/or biological reagent.
  • Chemical and/or biological reagents can be employed to protect and/or maintain the stability of the sample, including biomolecules (e.g., nucleic acid and protein) therein, during processing.
  • biomolecules e.g., nucleic acid and protein
  • One exemplary reagent is a protease inhibitor, which is generally used to protect or maintain the stability of protein during processing.
  • protease inhibitor which is generally used to protect or maintain the stability of protein during processing.
  • the methods, assays, and systems described herein can further comprise a step of obtaining or having obtained a test sample from a subject.
  • the subject can be a human subject.
  • the subject can be a subject in need of treatment for (e.g. having or diagnosed as having) premalignant lesions or a subject at risk of or at increased risk of developing bronchial premalignant lesions as described elsewhere herein.
  • the biopsy or brushing sample comprises morphologically-normal tissues or cells, e.g., the tissues or cells are not from a lesion and display normal morphology for their in vivo location.
  • the biopsy or brushing sample consists essentially of morphologically-normal tissues or cells.
  • the biopsy or brushing sample consists of morphologically-normal tissues or cells.
  • the biopsy or brushing sample comprises visually-normal tissues or cells, e.g., the tissues or cells are not from a lesion and to the unaided human eye have a normal appearance for their in vivo location.
  • the biopsy or brushing sample consists essentially of visually-normal tissues or cells.
  • the biopsy or brushing sample consists of visually-normal tissues or cells.
  • the biopsy or brushing sample comprises bronchial premalignant lesion cells. In some embodiments of any of the aspects, the biopsy or brushing sample consists essentially of bronchial premalignant lesion cells. In some embodiments of any of the aspects, the biopsy or brushing sample consists of bronchial premalignant lesion cells.
  • a level which is less than a reference level can be a level which is less by at least about 10%, at least about 20%, at least about 50%, at least about 60%, at least about 80%, at least about 90%, or less relative to the reference level. In some embodiments of any of the aspects, a level which is less than a reference level can be a level which is statistically significantly less than the reference level.
  • a level which is more than a reference level can be a level which is greater by at least about 10%, at least about 20%, at least about 50%, at least about 60%, at least about 80%, at least about 90%, at least about 100%, at least about 200%, at least about 300%, at least about 500% or more than the reference level.
  • a level which is more than a reference level can be a level which is statistically significantly greater than the reference level.
  • the reference can be a level of the target molecule in a population of subjects who do not have or are not diagnosed as having, and/or do not exhibit signs or symptoms of bronchial premalignant lesions. In some embodiments of any of the aspects, the reference can also be a level of expression of the target molecule in a control sample, a pooled sample of control individuals or a numeric value or range of values based on the same.
  • the reference can be the level of a target molecule in a sample obtained from the same subject at an earlier point in time, e.g., the methods described herein can be used to determine if a subject's sensitivity or response to a given therapy is changing over time or if the subtype of their lesions is changing.
  • the level of expression products of no more than 200 other genes is/are determined. In some embodiments of any of the aspects, the level of expression products of no more than 100 other genes is/are determined. In some embodiments of any of the aspects, the level of expression products of no more than 20 other genes is/are determined. In some embodiments of any of the aspects, the level of expression products of no more than 10 other genes is/are determined.
  • the expression level of a given gene can be normalized relative to the expression level of one or more reference genes or reference proteins.
  • the reference level can be the level in a sample of similar cell type, sample type, sample processing, and/or obtained from a subject of similar age, sex and other demographic parameters as the sample/subject for which the level of expression is to be determined.
  • the test sample and control reference sample are of the same type, that is, obtained from the same biological source, and comprising the same composition, e.g. the same number and type of cells.
  • the reference level can be a non-proliferative reference level, e.g., the level in a tissue or cell not comprising a proliferative lesion or from a subject who does not have a proliferative lesion.
  • the level can be the level in inflammatory, secretory, or normal-like lesion subtypes or an average or pooling thereof.
  • the methods described herein relate to treating a subject having or diagnosed as having bronchial premalignant lesions.
  • Subjects having bronchial premalignant lesions can be identified by a physician using current methods of diagnosing bronchial premalignant lesions.
  • Tests that may aid in a diagnosis of, e.g. bronchial premalignant lesions include, but are not limited to, bronchoscopy, autofluorescence bronchoscopy, etc.
  • a family history of bronchial premalignant lesions or exposure to risk factors for bronchial premalignant lesions e.g. cigarette smoke
  • compositions and methods described herein can be administered to a subject having or diagnosed as having bronchial premalignant lesions.
  • the methods described herein comprise administering an effective amount of compositions described herein to a subject in order to alleviate a symptom of a bronchial premalignant lesions.
  • “alleviating a symptom of a bronchial premalignant lesions” is ameliorating any condition or symptom associated with the bronchial premalignant lesions. As compared with an equivalent untreated control, such reduction is by at least 5%, 10%, 20%, 40%, 50%, 60%, 80%, 90%, 95%, 99% or more as measured by any standard technique.
  • compositions described herein can include, but are not limited to oral, parenteral, intravenous, intramuscular, subcutaneous, transdermal, airway (aerosol), pulmonary, cutaneous, topical, injection, or intratumoral administration. Administration can be local or systemic.
  • the methods described herein can prevent, delay, or slow the development of lung cancer, e.g., lung squamous cell carcinoma.
  • the subject treated according to the present methods is not a subject with lung cancer.
  • the subject treated according to the present methods is a subject who does not have lung cancer.
  • the subject treated according to the present methods is a subject who does not have and has not had lung cancer.
  • the subject treated according to the present methods is at risk of lung cancer.
  • the subject is a subject with a bronchial premalignant lesion.
  • the subject is a smoker. In some embodiments of any of the aspects, the subject is a former smoker. In some embodiments of any of the aspects, the subject is a non-smoker.
  • the treatments described herein e.g. an anti-proliferative drug, anti-inflammatory drug, or immune stimulating drug can be administered systemically, by inhalation, and/or topically to any portion of the airways of a subject (including the nose and mouth).
  • a treatment described herein, e.g. an anti-proliferative drug, anti-inflammatory drug, or immune stimulating drug can be administered i) systemically and ii) by inhalation or topically to any portion of the airways of a subject (including the nose and mouth) during a bronchoscopy or brushing collection.
  • An anti-proliferative drug is a drug that inhibits cell growth and/or division, e.g., cytostatic agents, wherein that is the primary activity of the compound in the relevant context.
  • Non-limiting examples of anti-proliferative drugs can include CDK inhibitors (e.g. purvalanol-a, palbociclib, ribociclib, abemaciclib, and olomoucine II); HDAC inhibitors (e.g. THM-I-94, vorinostat, givinostat); PARP inhibitors (e.g.
  • JAK inhibitors e.g.
  • JAK3-inhibitor-VI JAK3-inhibitor-VI, ruxolitinib, tofacitinib, oclacitinib, baricitinib, peficitinib, filgotinib, cerdulatinib, gandotinib, lestaurtinib, momelotinib, pacritinib, PF-04965842, upadacitinib, fedratinib, cucurbitacin, CHZ868); JNK inhibitors (e.g. ZG-10, AS-601245, AM-111); MTOR inhibitors (e.g.
  • AZD-8055 PI-103, rapamycin, temsirolimus, everolimus, ridaforolimus, rapalogs, sirolimus
  • FLT3 inhibitors e.g. lestaurtinib, TG-101348, gilteritinib, quizartinib, midostaurin, sorafenib, sunitinib
  • PI3K inhibitors e.g.
  • A-443644 pyrvinium-pamoate, VQD-002, perifosine, miltefosine, MK-2206, AZD5363, ipataseritib); tyrosine kinase inhibitors (e.g. aminopurvalanol-a, SU-11652, imatinib, gefitinib, erlotinib, sunitinib, adavosertib, lapatinib); protein kinase inhibitors (e.g.
  • HG-5-113-01 adavosertib, afatinib, axitinib, bosuntinib, cetuximab, conbimetinib, crizotinib, cabozantinib, dasatinib, entrectinib, erdafitinib, erlotinib, fostamatinib, gefitinib, ibrutinib, imatinib, lapatinib, lenvatinib, mubritinib, nilotinib, pazopanib, pegaptanib, ruxolitinib, sorafenib, sunitinib, SU6656, vandetanib, vemurafenib); RNA polymerase inhibitor (e.g.
  • topoisomerase inhibitors e.g. pidorubicine, doxorubicin, campothecins, indenosioquinolines, indotecan, imdimitecan, amsacrine, etoposide, teniposide, ICRF-193, genistein
  • HSP inhibitors e.g. HSP90-inhibitor, 17-N-Allylamino-17-demethoxygeldanamycin (17AAG), gamitrinib
  • DNA protein kinase inhibitors e.g., PIK-75
  • focal adhesion kinase inhibitors e.g.
  • anti-proliferative drugs include Acetylcholine receptor antagonists (e.g., clozapine, quetiapine, atropine, benztropine, biperiden, chlorpheniramine, citalopram, dycyclomine, dimenthydrinate, diphenhydramine, doxepin, doxylamine, glycopyrrolate, glycopyrronium, hyoscyamine, ipratropium, orphenadrine, oxitropium, oxybutynin, promethazine, propantheline bromide, scopolamine, solifenacin, solifenacin, tolterodine, tiotropium, trihexyphenidyl, tropicamide, tubocurarine, mecamylamine, hexamethonium, doxacurium, dextromethorphan, bupriopion); Acetylcholinesterase inhibitors (e.g., clozapine, quetiapin
  • Phosphodiesterase inhibitors e.g., vinpocetine, ENHA, BAY 60-7550, oxindole, PDP, IBMX, aminophylline, praxanthine, pentoxifylline, theobromine, inamrinone, milrinone, enoximone, anagrelide, cilostazol, pimobendan
  • SIRT inhibitors e.g., (s)-2-phentyl-6-chloro, 8-bormo-chroman-4-one, 3′-phenethyloxy-2-anilinobenzamide
  • sodium channel blockers e.g., procainamide, quinidine, disopyramide, lidocaine,
  • anti-proliferative drugs lacking anti-inflammatory activity in any context described herein can include JAK inhibitors, JNK inhibitors, AKT inhibitors, protein kinase inhibitors, RNA polymerase inhibitors, HSP inhibitors, DNA protein kinase inhibitors, focal adhesion inhibitors, RNA synthesis inhibitors, and mediator release inhibitors.
  • anti-inflammatory refers to a compound capable of reducing or inhibiting inflammation, wherein that is the primary activity of the compound in the relevant context.
  • anti-inflammatory drug or “anti-inflammatory agent” is used to describe any compound (including its analogs, derivatives, prodrugs and pharmaceutically salts) which can be used reduce or inhibit inflammation.
  • anti-inflammatory drugs can include NFkB pathway inhibitors (e.g.
  • anti-proliferative drugs include Acetylcholine receptor antagonist; Acetylcholinesterase inhibitors; Adenosine receptor antagonists; Adrenergic receptor antagonists; Angiotensin receptor antagonists; Apoptosis stimulants; Cyclooxygenase inhibitors; Cytokine production inhibitors; Dehydrogenase inhibitors; Dopamine receptor antagonist; EGFR inhibitors; ERK1 and ERK2 phosphorylation inhibitors; Estrogen receptor agonists; Glutamate receptor antagonists; Histamine receptor antagonists; Histone lysine methyltransferase inhibitors; IKK inhibitors; Ion channel antagonists; Leucine rich repeat kinase inhibitors; MDM inhibitors; Monoamine oxidase inhibitors; nucleophosmin inhibitors; PPAR receptor agonists; P
  • Non-examples of agents that can exhibit primarily an anti-inflammatory activity and/or an anti-proliferative activity, depending on the context can include Acetylcholine receptor antagonist, Acetylcholinesterase inhibitors, Adenosine receptor antagonists, Adrenergic receptor antagonists, Angiotensin receptor antagonists, Apoptosis stimulants, Aurora kinase inhibitors, CDK inhibitors, Cyclooxygenase inhibitors, Cytokine production inhibitors, Dehydrogenase inhibitors, Dopamine receptor antagonist, EGFR inhibitors, ERK1 and ERK2 phosphorylation inhibitors, Estrogen receptor agonists, FLT3 inhibitors, Glucocorticoid receptor agonists, Glutamate receptor antagonists, HDAC inhibitors, Histamine receptor antagonists, Histone lysine
  • an immune-stimulating drug is a drug that increases the activity of the immune system, preferably against cancer or dysplasia cells, wherein that is the primary activity of the compound in the relevant context.
  • the term “immune-stimulating drug” or “anti-inflammatory agent” is used to describe any compound (including its analogs, derivatives, prodrugs and pharmaceutically salts) which can be used stimulate the immune system.
  • immune stimulating drugs can include immune-checkpoint inhibitors (e.g. inhibitors against, PD-1, PD-L1, CTLA4, and LAG3); drugs that stimulate interferon signaling (e.g.
  • anti-viral drugs that improve interferon signaling
  • interferon signaling such as Pegintron, Pegasys, referon A, uniferon, multiferon, rebif, avonex, cinnovex, betaseron, actimmune, reiferon, pegetron
  • DNA synthesis inhibitors e.g., TAS-102, NC-6004, ganciclovir
  • CDK inhibitors e.g.
  • ribonucleotide reductase inhibitors e.g., motexafin, hydroxyurea, fludarabine, cladribine, gemcitabine, tezacitabine, triapine, gallium maltolate, gallium nitrate
  • dihydrofolate reductase inhibitors e.g., methotrexate, piritrexam, cycloguanil, JPC-2056
  • topoisomerase inhibitors e.g.
  • pidorubicine doxorubicin, campothecins, indenosioquinolines, indotecan, imdimitecan, amsacrine, etoposide, teniposide, ICRF-193, genistein); FLT3 inhibitors (e.g.
  • IGF-1 inhibitors IGF-1 inhibitors
  • MEK inhibitors e.g., trametinib, cobimetinib, binimetinib, selumetinib, PD-325901, TAK-733
  • aurora kinase inhibitors e.g., ZM447439, hesperidin, VX-680
  • PKC inhibitors e.g., ruboxistaurin, chelerythrine, miyabenol C, myricitrin, gossypol, verbascoside, BIM-1, bryostate 1, tamoxifen
  • RAF inhibitors e.g., vemurafenib, GDC-0879, PLX-4720, sorafenib, dabrafenib, LGX818); PDFGR
  • THM-I-94 vorinostat, givinostat
  • DNA methyltransferase inhibitors e.g., azacytidine, decitabine, zeublarine
  • EZH2 inhibitors DZNep, EPZ005687, EI1, GSK126, UNC1999, EPZ-6438, tazemetostat
  • immune stimulating drugs lacking anti-proliferative/inflammatory activity in any context described herein can include immune-checkpoint inhibitors (e.g. inhibitors against, PD-1, PD-L1, CTLA4, and LAG3); drugs that stimulate interferon signaling (e.g. anti-viral drugs that improve interferon signaling); DNA synthesis inhibitors; IMDH inhibitors; ribonucleotide reductase inhibitors; dihydrofolate reductase inhibitors; SRC inhibitors; retinoid receptor agonists; HDAC inhibitors; and DNA methyltransferase inhibitors.
  • immune-checkpoint inhibitors e.g. inhibitors against, PD-1, PD-L1, CTLA4, and LAG3
  • drugs that stimulate interferon signaling e.g. anti-viral drugs that improve interferon signaling
  • DNA synthesis inhibitors IMDH inhibitors
  • ribonucleotide reductase inhibitors ribonucleotide reductase inhibitor
  • an effective amount refers to the amount of a composition needed to alleviate at least one or more symptom of the disease or disorder, and relates to a sufficient amount of pharmacological composition to provide the desired effect.
  • the term “therapeutically effective amount” therefore refers to an amount of the composition that is sufficient to provide a particular therapeutic effect when administered to a typical subject.
  • An effective amount as used herein, in various contexts, would also include an amount sufficient to delay the development of a symptom of the disease, alter the course of a symptom disease (for example but not limited to, slowing the progression of a symptom of the disease), or reverse a symptom of the disease. Thus, it is not generally practicable to specify an exact “effective amount”. However, for any given case, an appropriate “effective amount” can be determined by one of ordinary skill in the art using only routine experimentation.
  • Effective amounts, toxicity, and therapeutic efficacy can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population).
  • the dosage can vary depending upon the dosage form employed and the route of administration utilized.
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and can be expressed as the ratio LD50/ED50.
  • Compositions and methods that exhibit large therapeutic indices are preferred.
  • a therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose can be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the active ingredient, which achieves a half-maximal inhibition of symptoms) as determined in cell culture, or in an appropriate animal model.
  • IC50 i.e., the concentration of the active ingredient, which achieves a half-maximal inhibition of symptoms
  • Levels in plasma can be measured, for example, by high performance liquid chromatography.
  • the effects of any particular dosage can be monitored by a suitable bioassay, e.g., assay for gene expression as described herein, among others.
  • the dosage can be determined by a physician and adjusted, as necessary, to suit observed effects of the treatment.
  • the technology described herein relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a drug as described herein, and optionally a pharmaceutically acceptable carrier.
  • the active ingredients of the pharmaceutical composition comprise the drug as described herein.
  • the active ingredients of the pharmaceutical composition consist essentially of the drug as described herein.
  • the active ingredients of the pharmaceutical composition consist of the drug as described herein.
  • Pharmaceutically acceptable carriers and diluents include saline, aqueous buffer solutions, solvents and/or dispersion media. The use of such carriers and diluents is well known in the art.
  • materials which can serve as pharmaceutically-acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, methylcellulose, ethyl cellulose, microcrystalline cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) lubricating agents, such as magnesium stearate, sodium lauryl sulfate and talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol (PEG); (12) esters, such as e
  • wetting agents, coloring agents, release agents, coating agents, sweetening agents, flavoring agents, perfuming agents, preservative and antioxidants can also be present in the formulation.
  • the terms such as “excipient”, “carrier”, “pharmaceutically acceptable carrier” or the like are used interchangeably herein.
  • the carrier inhibits the degradation of the active agent.
  • the pharmaceutical composition comprising a drug as described herein can be a parenteral dose form. Since administration of parenteral dosage forms typically bypasses the patient's natural defenses against contaminants, parenteral dosage forms are preferably sterile or capable of being sterilized prior to administration to a patient. Examples of parenteral dosage forms include, but are not limited to, solutions ready for injection, dry products ready to be dissolved or suspended in a pharmaceutically acceptable vehicle for injection, suspensions ready for injection, and emulsions. In addition, controlled-release parenteral dosage forms can be prepared for administration of a patient, including, but not limited to, DUROS®-type dosage forms and dose-dumping.
  • Suitable vehicles that can be used to provide parenteral dosage forms of a drug as disclosed within are well known to those skilled in the art. Examples include, without limitation: sterile water; water for injection USP; saline solution; glucose solution; aqueous vehicles such as but not limited to, sodium chloride injection, Ringer's injection, dextrose Injection, dextrose and sodium chloride injection, and lactated Ringer's injection; water-miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glycol, and propylene glycol; and non-aqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.
  • Compounds that alter or modify the solubility of a pharmaceutically acceptable salt of the drug as disclosed herein can also be incorporated into the parenteral dosage forms of the disclosure, including conventional and controlled-release parenteral dosage
  • compositions comprising a drug can also be formulated to be suitable for oral administration, for example as discrete dosage forms, such as, but not limited to, tablets (including without limitation scored or coated tablets), pills, caplets, capsules, chewable tablets, powder packets, cachets, troches, wafers, aerosol sprays, or liquids, such as but not limited to, syrups, elixirs, solutions or suspensions in an aqueous liquid, a non-aqueous liquid, an oil-in-water emulsion, or a water-in-oil emulsion.
  • Such compositions contain a predetermined amount of the pharmaceutically acceptable salt of the disclosed compounds, and may be prepared by methods of pharmacy well known to those skilled in the art. See generally, Remington: The Science and Practice of Pharmacy, 21st Ed., Lippincott, Williams, and Wilkins, Philadelphia Pa. (2005).
  • Conventional dosage forms generally provide rapid or immediate drug release from the formulation. Depending on the pharmacology and pharmacokinetics of the drug, use of conventional dosage forms can lead to wide fluctuations in the concentrations of the drug in a patient's blood and other tissues. These fluctuations can impact a number of parameters, such as dose frequency, onset of action, duration of efficacy, maintenance of therapeutic blood levels, toxicity, side effects, and the like.
  • controlled-release formulations can be used to control a drug's onset of action, duration of action, plasma levels within the therapeutic window, and peak blood levels.
  • controlled- or extended-release dosage forms or formulations can be used to ensure that the maximum effectiveness of a drug is achieved while minimizing potential adverse effects and safety concerns, which can occur both from under-dosing a drug (i.e., going below the minimum therapeutic levels) as well as exceeding the toxicity level for the drug.
  • the drug can be administered in a sustained release formulation.
  • Controlled-release pharmaceutical products have a common goal of improving drug therapy over that achieved by their non-controlled release counterparts.
  • the use of an optimally designed controlled-release preparation in medical treatment is characterized by a minimum of drug substance being employed to cure or control the condition in a minimum amount of time.
  • Advantages of controlled-release formulations include: 1) extended activity of the drug; 2) reduced dosage frequency; 3) increased patient compliance; 4) usage of less total drug; 5) reduction in local or systemic side effects; 6) minimization of drug accumulation; 7) reduction in blood level fluctuations; 8) improvement in efficacy of treatment; 9) reduction of potentiation or loss of drug activity; and 10) improvement in speed of control of diseases or conditions. Kim, Cherng-ju, Controlled Release Dosage Form Design, 2 (Technomic Publishing, Lancaster, Pa.: 2000).
  • Controlled-release formulations are designed to initially release an amount of drug (active ingredient) that promptly produces the desired therapeutic effect, and gradually and continually release other amounts of drug to maintain this level of therapeutic or prophylactic effect over an extended period of time. In order to maintain this constant level of drug in the body, the drug must be released from the dosage form at a rate that will replace the amount of drug being metabolized and excreted from the body.
  • Controlled-release of an active ingredient can be stimulated by various conditions including, but not limited to, pH, ionic strength, osmotic pressure, temperature, enzymes, water, and other physiological conditions or compounds.
  • a variety of known controlled- or extended-release dosage forms, formulations, and devices can be adapted for use with the salts and compositions of the disclosure. Examples include, but are not limited to, those described in U.S. Pat. Nos. 3,845,770; 3,916,899; 3,536,809; 3,598,123; 4,008,719; 5674,533; 5,059,595; 5,591,767; 5,120,548; 5,073,543; 5,639,476; 5,354,556; 5,733,566; and 6,365,185 B1; each of which is incorporated herein by reference.
  • dosage forms can be used to provide slow or controlled-release of one or more active ingredients using, for example, hydroxypropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems (such as OROS® (Alza Corporation, Mountain View, Calif. USA)), or a combination thereof to provide the desired release profile in varying proportions.
  • active ingredients for example, hydroxypropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems (such as OROS® (Alza Corporation, Mountain View, Calif. USA)), or a combination thereof to provide the desired release profile in varying proportions.
  • OROS® Alza Corporation, Mountain View, Calif. USA
  • the drug described herein is administered as a monotherapy, e.g., another treatment for the bronchial premalignant lesions is not administered to the subject.
  • the methods described herein can further comprise administering a second agent and/or treatment to the subject, e.g. as part of a combinatorial therapy.
  • an effective dose of a composition comprising a drug as described herein can be administered to a patient once.
  • an effective dose of a composition comprising a drug can be administered to a patient repeatedly.
  • subjects can be administered a therapeutic amount of a composition comprising a drug, such as, e.g. 0.1 mg/kg, 0.5 mg/kg, 1.0 mg/kg, 2.0 mg/kg, 2.5 mg/kg, 5 mg/kg, 10 mg/kg, 15 mg/kg, 20 mg/kg, 25 mg/kg, 30 mg/kg, 40 mg/kg, 50 mg/kg, or more.
  • the treatments can be administered on a less frequent basis. For example, after treatment biweekly for three months, treatment can be repeated once per month, for six months or a year or longer.
  • Treatment according to the methods described herein can reduce levels of a marker or symptom of a condition, e.g. by at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80% or at least 90% or more.
  • the dosage of a composition as described herein can be determined by a physician and adjusted, as necessary, to suit observed effects of the treatment. With respect to duration and frequency of treatment, it is typical for skilled clinicians to monitor subjects in order to determine when the treatment is providing therapeutic benefit, and to determine whether to increase or decrease dosage, increase or decrease administration frequency, discontinue treatment, resume treatment, or make other alterations to the treatment regimen.
  • the dosing schedule can vary from once a week to daily depending on a number of clinical factors, such as the subject's sensitivity to the active ingredient.
  • the desired dose or amount of activation can be administered at one time or divided into subdoses, e.g., 2-4 subdoses and administered over a period of time, e.g., at appropriate intervals through the day or other appropriate schedule.
  • administration can be chronic, e.g., one or more doses and/or treatments daily over a period of weeks or months.
  • dosing and/or treatment schedules are administration daily, twice daily, three times daily or four or more times daily over a period of 1 week, 2 weeks, 3 weeks, 4 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, or 6 months, or more.
  • a composition comprising a drug described herein can be administered over a period of time, such as over a 5 minute, 10 minute, 15 minute, 20 minute, or 25 minute period.
  • the dosage ranges for the administration of a drug, according to the methods described herein depend upon, for example, the form of the drug, its potency, and the extent to which symptoms, markers, or indicators of a condition described herein are desired to be reduced, for example the percentage reduction desired for lesion size or the extent to which, for example, lesion subtype changes are desired to be induced.
  • the dosage should not be so large as to cause adverse side effects.
  • the dosage will vary with the age, condition, and sex of the patient and can be determined by one of skill in the art.
  • the dosage can also be adjusted by the individual physician in the event of any complication.
  • the efficacy of a drug in, e.g. the treatment of a condition described herein, or to induce a response as described herein (e.g. reduction in lesion size) can be determined by the skilled clinician.
  • a treatment is considered “effective treatment,” as the term is used herein, if one or more of the signs or symptoms of a condition described herein are altered in a beneficial manner, other clinically accepted symptoms are improved, or even ameliorated, or a desired response is induced e.g., by at least 10% following treatment according to the methods described herein.
  • Efficacy can be assessed, for example, by measuring a marker, indicator, symptom, and/or the incidence of a condition treated according to the methods described herein or any other measurable parameter appropriate.
  • Treatment includes any treatment of a disease in an individual or an animal (some non-limiting examples include a human or an animal) and includes: (1) inhibiting the disease, e.g., preventing a worsening of symptoms (e.g. pain or inflammation); or (2) relieving the severity of the disease, e.g., causing regression of symptoms.
  • An effective amount for the treatment of a disease means that amount which, when administered to a subject in need thereof, is sufficient to result in effective treatment as that term is defined herein, for that disease.
  • Efficacy of an agent can be determined by assessing physical indicators of a condition or desired response. It is well within the ability of one skilled in the art to monitor efficacy of administration and/or treatment by measuring any one of such parameters, or any combination of parameters. Efficacy can be assessed in animal models of a condition described herein, for example treatment of a mouse model of bronchial premalignant lesions. When using an experimental animal model, efficacy of treatment is evidenced when a statistically significant change in a marker is observed, e.g. lesion size or gene expression.
  • a bronchoscopy-based procedure refers to any endoscopic technique that permits examination of the bronchus and/or lungs.
  • Bronchoscopy-based procedures can include white light bronchoscopy, autofluorescence bronchoscopy, flexible bronchoscopy, rigid bronchoscopy, bronchoalveolar lavage, and the like. Bronchoscopy-based procedures can further include biopsy, brushing, or tissue sampling. If the
  • the methods and biomarker signatures described herein can be applied to methods of predicting the risk of lung cancer in a subject and/or determining the efficacy of treatment or need for further treatment. For example, transition from a proliferative or inflammatory subtype to a normal-like or secretory subtype would indicate that a treatment had been effective or that the treatment can be discontinued.
  • a method of predicting the risk, or the likelihood of progression to lung cancer in a subject comprising: detecting the level of expression of at least one module 5 gene and/or at least one module 6 gene in a sample obtained from the subject, wherein an increased level of expression of at least one module 5 gene as compared to a non-proliferative lesion reference level; and/or a decreased level of expression of at least one module 6 gene as compared to a non-proliferative lesion reference level indicates an increased risk of or likelihood of progressing to lung cancer.
  • a method of predicting the risk, or the likelihood of progression to lung cancer in a subject comprising: detecting the level of expression of at least one module 5 gene and/or at least one module 6 gene in a sample obtained from the subject at a first time point, and detecting the level of expression of at least one module 5 gene and/or at least one module 6 gene in a sample obtained from the subject at a second, subsequent time point, wherein an increased level of expression of at least one module 5 gene over time; and/or a decreased level of expression of at least one module 6 gene over time indicates an increased risk of or likelihood of progressing to lung cancer.
  • a method of predicting the risk, or the likelihood of progression to lung cancer in a subject comprising: detecting the level of expression of at least one module 9 gene and/or at least one module 10 gene in a sample obtained from the subject, wherein an increased level of expression of at least one module 10 gene as compared to a non-proliferative lesion reference level; and/or a decreased level of expression of at least one module 9 gene as compared to a non-proliferative lesion reference level indicates an increased risk of or likelihood of progressing to lung cancer.
  • a method of predicting the risk, or the likelihood of progression to lung cancer in a subject comprising: detecting the level of expression of at least one module 10 gene and/or at least one module 9 gene in a sample obtained from the subject at a first time point, and detecting the level of expression of at least one module 9 gene and/or at least one module 10 gene in a sample obtained from the subject at a second, subsequent time point, wherein an increased level of expression of at least one module 10 gene over time; and/or a decreased level of expression of at least one module 9 gene over time indicates an increased risk of or likelihood of progressing to lung cancer.
  • a method of predicting the risk, or the likelihood of progression to lung cancer in a subject comprising: detecting the level of expression of at least one module 2 gene and/or at least one module 6 gene in a sample obtained from the subject, wherein an increased level of expression of at least one module 2 gene as compared to a non-proliferative lesion reference level; and/or a decreased level of expression of at least one module 6 gene as compared to a non-proliferative lesion reference level indicates an increased risk of or likelihood of progressing to lung cancer.
  • a method of predicting the risk, or the likelihood of progression to lung cancer in a subject comprising: detecting the level of expression of at least one module 2 gene and/or at least one module 6 gene in a sample obtained from the subject at a first time point, and detecting the level of expression of at least one module 2 gene and/or at least one module 6 gene in a sample obtained from the subject at a second, subsequent time point, wherein an increased level of expression of at least one module 2 gene over time; and/or a decreased level of expression of at least one module 6 gene over time indicates an increased risk of or likelihood of progressing to lung cancer.
  • a method of determining treatment efficacy comprising: detecting the level of expression of at least one module 5 gene and/or at least one module 6 gene in a sample obtained from the subject at a first time point, administering a treatment or candidate treatment, and detecting the level of expression of at least one module 5 gene and/or at least one module 6 gene in a sample obtained from the subject at a second, subsequent time point, wherein an decreased level of expression of at least one module 5 gene over time; and/or an increased level of expression of at least one module 6 gene over time indicates the treatment is effective.
  • a method of treatment efficacy comprising: detecting the level of expression of at least one module 10 gene and/or at least one module 9 gene in a sample obtained from the subject at a first time point, administering a treatment or candidate treatment, and detecting the level of expression of at least one module 9 gene and/or at least one module 10 gene in a sample obtained from the subject at a second, subsequent time point, wherein an decreased level of expression of at least one module 10 gene over time; and/or an increased level of expression of at least one module 9 gene over time indicates the treatment is effective.
  • a method of determining treatment efficacy comprising: detecting the level of expression of at least one module 2 gene and/or at least one module 6 gene in a sample obtained from the subject at a first time point, administering a treatment or candidate treatment, and detecting the level of expression of at least one module 2 gene and/or at least one module 6 gene in a sample obtained from the subject at a second, subsequent time point, wherein an decreased level of expression of at least one module 2 gene over time; and/or an increased level of expression of at least one module 6 gene over time indicates the treatment is effective.
  • a method comprising: detecting the level of expression of at least one module 5 gene and/or at least one module 6 gene in a sample obtained from a subject, wherein the level of expression of no more than 1,000 (e.g., no more than 500, 400, 300, 200, or 100) genes is determined.
  • a method comprising: detecting the level of expression of at least one module 9 gene and/or at least one module 10 gene in a sample obtained from a subject, wherein the level of expression of no more than 1,000 (e.g., no more than 500, 400, 300, 200, or 100) genes is determined.
  • a method comprising: detecting the level of expression of at least one module 2 gene and/or at least one module 6 gene in a sample obtained from a subject, wherein the level of expression of no more than 1,000 (e.g., no more than 500, 400, 300, 200, or 100) genes is determined.
  • the sample is a bronchial brushing sample.
  • the at least one gene is selected from Table 14 or 15.
  • the at least one gene can be one or more genes selected from Tables 13, 14, 15, and/or 16.
  • the gene lists of Tables 13, 14 and 16 are relevant to endobronchial biopsy samples that range in histology from normal to premalignant.
  • the sample is an endobronchial biopsy sample
  • the one or more genes are selected from Table 13, 14 and/or 16.
  • Table 15 is relevant for normal bronchial brushings.
  • the sample is bronchial brushing sample (e.g, of normal tissue)
  • the one or more genes are selected from Table 15.
  • the one or more genes selected from Table 13 or 16 are not B2M, HLA-DRA, HLA-DRB1, or HLA-DPA 1. In some embodiments of any of the aspects, if the one or more genes selected from Table 13 or 16 include B2M, HLA-DRA, HLA-DRB1, and/or HLA-DPA1, at least one additional gene from Table 13 or 16 is selected.
  • cancer relates generally to a class of diseases or conditions in which abnormal cells divide without control and can invade nearby tissues. Cancer cells can also spread to other parts of the body through the blood and lymph systems.
  • Carcinoma is a cancer that begins in the skin or in tissues that line or cover internal organs.
  • Sarcoma is a cancer that begins in bone, cartilage, fat, muscle, blood vessels, or other connective or supportive tissue.
  • Leukemia is a cancer that starts in blood-forming tissue such as the bone marrow, and causes large numbers of abnormal blood cells to be produced and enter the blood.
  • Lymphoma and multiple myeloma are cancers that begin in the cells of the immune system.
  • Central nervous system cancers are cancers that begin in the tissues of the brain and spinal cord.
  • the cancer is a primary cancer. In some embodiments of any of the aspects, the cancer is a malignant cancer.
  • malignant refers to a cancer in which a group of tumor cells display one or more of uncontrolled growth (i.e., division beyond normal limits), invasion (i.e., intrusion on and destruction of adjacent tissues), and metastasis (i.e., spread to other locations in the body via lymph or blood).
  • metastasize refers to the spread of cancer from one part of the body to another.
  • a tumor formed by cells that have spread is called a “metastatic tumor” or a “metastasis.”
  • the metastatic tumor contains cells that are like those in the original (primary) tumor.
  • the term “benign” or “non-malignant” refers to tumors that may grow larger but do not spread to other parts of the body. Benign tumors are self-limited and typically do not invade or metastasize.
  • a “cancer cell” or “tumor cell” refers to an individual cell of a cancerous growth or tissue.
  • a tumor refers generally to a swelling or lesion formed by an abnormal growth of cells, which may be benign, pre-malignant, or malignant. Most cancer cells form tumors, but some, e.g., leukemia, do not necessarily form tumors. For those cancer cells that form tumors, the terms cancer (cell) and tumor (cell) are used interchangeably.
  • neoplasm refers to any new and abnormal growth of tissue, e.g., an abnormal mass of tissue, the growth of which exceeds and is uncoordinated with that of the normal tissues.
  • a neoplasm can be a benign neoplasm, premalignant neoplasm, or a malignant neoplasm.
  • a subject that has a cancer or a tumor is a subject having objectively measurable cancer cells present in the subject's body. Included in this definition are malignant, actively proliferative cancers, as well as potentially dormant tumors or micrometastatses. Cancers which migrate from their original location and seed other vital organs can eventually lead to the death of the subject through the functional deterioration of the affected organs.
  • cancer examples include but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, leukemia, basal cell carcinoma, biliary tract cancer; bladder cancer; bone cancer; brain and CNS cancer; breast cancer; cancer of the peritoneum; cervical cancer; choriocarcinoma; colon and rectum cancer; connective tissue cancer; cancer of the digestive system; endometrial cancer; esophageal cancer; eye cancer; cancer of the head and neck; gastric cancer (including gastrointestinal cancer); glioblastoma (GBM); hepatic carcinoma; hepatoma; intra-epithelial neoplasm.; kidney or renal cancer; larynx cancer; leukemia; liver cancer; lung cancer (e.g., small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, and squamous carcinoma of the lung); lymphoma including Hodgkin's and non-Hodgkin's lymphoma; mel
  • a “cancer cell” is a cancerous, pre-cancerous, or transformed cell, either in vivo, ex vivo, or in tissue culture, that has spontaneous or induced phenotypic changes that do not necessarily involve the uptake of new genetic material.
  • transformation can arise from infection with a transforming virus and incorporation of new genomic nucleic acid, or uptake of exogenous nucleic acid, it can also arise spontaneously or following exposure to a carcinogen, thereby mutating an endogenous gene.
  • Transformation/cancer is associated with, e.g., morphological changes, immortalization of cells, aberrant growth control, foci formation, anchorage independence, malignancy, loss of contact inhibition and density limitation of growth, growth factor or serum independence, tumor specific markers, invasiveness or metastasis, and tumor growth in suitable animal hosts such as nude mice.
  • “decrease”, “reduced”, “reduction”, or “inhibit” are all used herein to mean a decrease by a statistically significant amount. In some embodiments, “reduce,” “reduction” or “decrease” or “inhibit” typically means a decrease by at least 10% as compared to a reference level (e.g.
  • “reduction” or “inhibition” does not encompass a complete inhibition or reduction as compared to a reference level.
  • “Complete inhibition” is a 100% inhibition as compared to a reference level.
  • a decrease can be preferably down to a level accepted as within the range of normal for an individual without a given disorder.
  • the terms “increased”, “increase”, “enhance”, or “activate” are all used herein to mean an increase by a statically significant amount.
  • the terms “increased”, “increase”, “enhance”, or “activate” can mean an increase of at least 10% as compared to a reference level, for example an increase of at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or up to and including a 100% increase or any increase between 10-100% as compared to a reference level, or at least about a 2-fold, or at least about a 3-fold, or at least about a 4-fold, or at least about a 5-fold or at least about a 10-fold increase, or any increase between 2-fold and 10-fold or greater as compared to a reference level.
  • a “increase” is a statistically significant increase in such level.
  • a “subject” means a human or animal. Usually the animal is a vertebrate such as a primate, rodent, domestic animal or game animal. Primates include chimpanzees, cynomologous monkeys, spider monkeys, and macaques, e.g., Rhesus. Rodents include mice, rats, woodchucks, ferrets, rabbits and hamsters.
  • Domestic and game animals include cows, horses, pigs, deer, bison, buffalo, feline species, e.g., domestic cat, canine species, e.g., dog, fox, wolf, avian species, e.g., chicken, emu, ostrich, and fish, e.g., trout, catfish and salmon.
  • the subject is a mammal, e.g., a primate, e.g., a human.
  • the terms, “individual,” “patient” and “subject” are used interchangeably herein.
  • the subject is a mammal.
  • the mammal can be a human, non-human primate, mouse, rat, dog, cat, horse, or cow, but is not limited to these examples. Mammals other than humans can be advantageously used as subjects that represent animal models of bronchial premalignant lesions.
  • a subject can be male or female.
  • a subject can be one who has been previously diagnosed with or identified as suffering from or having a condition in need of treatment (e.g. bronchial premalignant lesions) or one or more complications related to such a condition, and optionally, have already undergone treatment for bronchial premalignant lesions or the one or more complications related to bronchial premalignant lesions.
  • a subject can also be one who has not been previously diagnosed as having bronchial premalignant lesions or one or more complications related to bronchial premalignant lesions.
  • a subject can be one who exhibits one or more risk factors for bronchial premalignant lesions or one or more complications related to bronchial premalignant lesions or a subject who does not exhibit risk factors.
  • a “subject in need” of treatment for a particular condition can be a subject having that condition, diagnosed as having that condition, or at risk of developing that condition.
  • protein and “polypeptide” are used interchangeably herein to designate a series of amino acid residues, connected to each other by peptide bonds between the alpha-amino and carboxy groups of adjacent residues.
  • protein and “polypeptide” refer to a polymer of amino acids, including modified amino acids (e.g., phosphorylated, glycated, glycosylated, etc.) and amino acid analogs, regardless of its size or function.
  • Protein and “polypeptide” are often used in reference to relatively large polypeptides, whereas the term “peptide” is often used in reference to small polypeptides, but usage of these terms in the art overlaps.
  • polypeptide proteins and “polypeptide” are used interchangeably herein when referring to a gene product and fragments thereof.
  • exemplary polypeptides or proteins include gene products, naturally occurring proteins, homologs, orthologs, paralogs, fragments and other equivalents, variants, fragments, and analogs of the foregoing.
  • variants naturally occurring or otherwise
  • alleles homologs
  • conservatively modified variants conservative substitution variants of any of the particular polypeptides described are encompassed.
  • amino acid sequences one of skill will recognize that individual substitutions, deletions or additions to a nucleic acid, peptide, polypeptide, or protein sequence which alters a single amino acid or a small percentage of amino acids in the encoded sequence is a “conservatively modified variant” where the alteration results in the substitution of an amino acid with a chemically similar amino acid and retains the desired activity of the polypeptide.
  • conservatively modified variants are in addition to and do not exclude polymorphic variants, interspecies homologs, and alleles consistent with the disclosure.
  • a given amino acid can be replaced by a residue having similar physiochemical characteristics, e.g., substituting one aliphatic residue for another (such as Ile, Val, Leu, or Ala for one another), or substitution of one polar residue for another (such as between Lys and Arg; Glu and Asp; or Gln and Asn).
  • Other such conservative substitutions e.g., substitutions of entire regions having similar hydrophobicity characteristics, are well known.
  • Polypeptides comprising conservative amino acid substitutions can be tested in any one of the assays described herein to confirm that a desired activity, e.g. activity and specificity of a native or reference polypeptide is retained.
  • Amino acids can be grouped according to similarities in the properties of their side chains (in A. L. Lehninger, in Biochemistry, second ed., pp. 73-75, Worth Publishers, New York (1975)): (1) non-polar: Ala (A), Val (V), Leu (L), Ile (I), Pro (P), Phe (F), Trp (W), Met (M); (2) uncharged polar: Gly (G), Ser (S), Thr (T), Cys (C), Tyr (Y), Asn (N), Gln (Q); (3) acidic: Asp (D), Glu (E); (4) basic: Lys (K), Arg (R), His (H).
  • Naturally occurring residues can be divided into groups based on common side-chain properties: (1) hydrophobic: Norleucine, Met, Ala, Val, Leu, Ile; (2) neutral hydrophilic: Cys, Ser, Thr, Asn, Gln; (3) acidic: Asp, Glu; (4) basic: His, Lys, Arg; (5) residues that influence chain orientation: Gly, Pro; (6) aromatic: Trp, Tyr, Phe.
  • Non-conservative substitutions will entail exchanging a member of one of these classes for another class.
  • Particular conservative substitutions include, for example; Ala into Gly or into Ser; Arg into Lys; Asn into Gln or into His; Asp into Glu; Cys into Ser; Gln into Asn; Glu into Asp; Gly into Ala or into Pro; His into Asn or into Gln; Ile into Leu or into Val; Leu into Ile or into Val; Lys into Arg, into Gln or into Glu; Met into Leu, into Tyr or into Ile; Phe into Met, into Leu or into Tyr; Ser into Thr; Thr into Ser; Trp into Tyr; Tyr into Trp; and/or Phe into Val, into Ile or into Leu.
  • the polypeptide described herein can be a functional fragment of one of the amino acid sequences described herein.
  • a “functional fragment” is a fragment or segment of a peptide which retains at least 50% of the wildtype reference polypeptide's activity according to the assays described below herein.
  • a functional fragment can comprise conservative substitutions of the sequences disclosed herein.
  • the polypeptide described herein can be a variant of a sequence described herein.
  • the variant is a conservatively modified variant.
  • Conservative substitution variants can be obtained by mutations of native nucleotide sequences, for example.
  • a “variant,” as referred to herein, is a polypeptide substantially homologous to a native or reference polypeptide, but which has an amino acid sequence different from that of the native or reference polypeptide because of one or a plurality of deletions, insertions or substitutions.
  • Variant polypeptide-encoding DNA sequences encompass sequences that comprise one or more additions, deletions, or substitutions of nucleotides when compared to a native or reference DNA sequence, but that encode a variant protein or fragment thereof that retains activity.
  • a wide variety of PCR-based site-specific mutagenesis approaches are known in the art and can be applied by the ordinarily skilled artisan.
  • a variant amino acid or DNA sequence can be at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or more, identical to a native or reference sequence.
  • the degree of homology (percent identity) between a native and a mutant sequence can be determined, for example, by comparing the two sequences using freely available computer programs commonly employed for this purpose on the world wide web (e.g. BLASTp or BLASTn with default settings).
  • Alterations of the native amino acid sequence can be accomplished by any of a number of techniques known to one of skill in the art. Mutations can be introduced, for example, at particular loci by synthesizing oligonucleotides containing a mutant sequence, flanked by restriction sites enabling ligation to fragments of the native sequence. Following ligation, the resulting reconstructed sequence encodes an analog having the desired amino acid insertion, substitution, or deletion. Alternatively, oligonucleotide-directed site-specific mutagenesis procedures can be employed to provide an altered nucleotide sequence having particular codons altered according to the substitution, deletion, or insertion required. Techniques for making such alterations are very well established and include, for example, those disclosed by Walder et al.
  • Any cysteine residue not involved in maintaining the proper conformation of the polypeptide also can be substituted, generally with serine, to improve the oxidative stability of the molecule and prevent aberrant crosslinking. Conversely, cysteine bond(s) can be added to the polypeptide to improve its stability or facilitate oligomerization.
  • nucleic acid or “nucleic acid sequence” refers to any molecule, preferably a polymeric molecule, incorporating units of ribonucleic acid, deoxyribonucleic acid or an analog thereof.
  • the nucleic acid can be either single-stranded or double-stranded.
  • a single-stranded nucleic acid can be one nucleic acid strand of a denatured double-stranded DNA. Alternatively, it can be a single-stranded nucleic acid not derived from any double-stranded DNA.
  • the nucleic acid can be DNA.
  • nucleic acid can be RNA.
  • Suitable DNA can include, e.g., genomic DNA or cDNA.
  • Suitable RNA can include, e.g., mRNA.
  • expression refers to the cellular processes involved in producing RNA and proteins and as appropriate, secreting proteins, including where applicable, but not limited to, for example, transcription, transcript processing, translation and protein folding, modification and processing.
  • Expression can refer to the transcription and stable accumulation of sense (mRNA) or antisense RNA derived from a nucleic acid fragment or fragments of the invention and/or to the translation of mRNA into a polypeptide.
  • the expression of a biomarker(s), target(s), or gene/polypeptide described herein is/are tissue-specific. In some embodiments, the expression of a biomarker(s), target(s), or gene/polypeptide described herein is/are global. In some embodiments, the expression of a biomarker(s), target(s), or gene/polypeptide described herein is systemic.
  • “Expression products” include RNA transcribed from a gene, and polypeptides obtained by translation of mRNA transcribed from a gene.
  • the term “gene” means the nucleic acid sequence which is transcribed (DNA) to RNA in vitro or in vivo when operably linked to appropriate regulatory sequences.
  • the gene may or may not include regions preceding and following the coding region, e.g. 5′ untranslated (5′UTR) or “leader” sequences and 3′ UTR or “trailer” sequences, as well as intervening sequences (introns) between individual coding segments (exons).
  • Marker in the context of the present invention refers to an expression product, e.g., nucleic acid or polypeptide which is differentially present in a sample taken from subjects having bronchial premalignant lesions of a particular subtype, as compared to a comparable sample taken from control subjects (e.g., a healthy subject).
  • biomarker is used interchangeably with the term “marker.”
  • the methods described herein relate to measuring, detecting, or determining the level of at least one marker.
  • detecting or “measuring” refers to observing a signal from, e.g. a probe, label, or target molecule to indicate the presence of an analyte in a sample. Any method known in the art for detecting a particular label moiety can be used for detection. Exemplary detection methods include, but are not limited to, spectroscopic, fluorescent, photochemical, biochemical, immunochemical, electrical, optical or chemical methods. In some embodiments of any of the aspects, measuring can be a quantitative observation.
  • the terms “treat,” “treatment,” “treating,” or “amelioration” refer to therapeutic treatments, wherein the object is to reverse, alleviate, ameliorate, inhibit, slow down or stop the progression or severity of a condition associated with a disease or disorder, e.g. bronchial premalignant lesion.
  • the term “treating” includes reducing or alleviating at least one adverse effect or symptom of a condition, disease or disorder associated with a bronchial premalignant lesion.
  • Treatment is generally “effective” if one or more symptoms or clinical markers are reduced. Alternatively, treatment is “effective” if the progression of a disease is reduced or halted.
  • treatment includes not just the improvement of symptoms or markers, but also a cessation of, or at least slowing of, progress or worsening of symptoms compared to what would be expected in the absence of treatment.
  • Beneficial or desired clinical results include, but are not limited to, alleviation of one or more symptom(s), diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, remission (whether partial or total), and/or decreased mortality, whether detectable or undetectable.
  • treatment also includes providing relief from the symptoms or side-effects of the disease (including palliative treatment).
  • the term “pharmaceutical composition” refers to the active agent in combination with a pharmaceutically acceptable carrier e.g. a carrier commonly used in the pharmaceutical industry.
  • a pharmaceutically acceptable carrier e.g. a carrier commonly used in the pharmaceutical industry.
  • pharmaceutically acceptable is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • a pharmaceutically acceptable carrier can be a carrier other than water.
  • a pharmaceutically acceptable carrier can be a cream, emulsion, gel, liposome, nanoparticle, and/or ointment.
  • a pharmaceutically acceptable carrier can be an artificial or engineered carrier, e.g., a carrier that the active ingredient would not be found to occur in nature.
  • administering refers to the placement of a compound as disclosed herein into a subject by a method or route which results in at least partial delivery of the agent at a desired site.
  • Pharmaceutical compositions comprising the compounds disclosed herein can be administered by any appropriate route which results in an effective treatment in the subject.
  • administration comprises physical human activity, e.g., an injection, act of ingestion, an act of application, and/or manipulation of a delivery device or machine. Such activity can be performed, e.g., by a medical professional and/or the subject being treated.
  • contacting refers to any suitable means for delivering, or exposing, an agent to at least one cell.
  • exemplary delivery methods include, but are not limited to, direct delivery to cell culture medium, perfusion, injection, or other delivery method well known to one skilled in the art.
  • contacting comprises physical human activity, e.g., an injection; an act of dispensing, mixing, and/or decanting; and/or manipulation of a delivery device or machine.
  • inhibitor refers to an agent which can decrease the expression and/or activity of the target molecule or activity or process, e.g. by at least 10% or more, e.g. by 10% or more, 50% or more, 70% or more, 80% or more, 90% or more, 95% or more, or 98% or more.
  • drug As used herein, the terms “drug”, “compound” or “agent” are used interchangeably and refer to molecules and/or compositions.
  • the compounds/agents include, but are not limited to, chemical compounds and mixtures of chemical compounds, e.g., small organic or inorganic molecules; saccharines; oligosaccharides; polysaccharides; biological macromolecules, e.g., peptides, proteins, and peptide analogs and derivatives; peptidomimetics; nucleic acids; nucleic acid analogs and derivatives; extracts made from biological materials such as bacteria, plants, fungi, or animal cells or tissues; naturally occurring or synthetic compositions; peptides; aptamers; and antibodies and intrabodies, or fragments thereof.
  • “drug” as used herein refers to an agent approved for medical use, e.g., by the FDA.
  • statically significant or “significantly” refers to statistical significance and generally means a two standard deviation (2SD) or greater difference.
  • compositions, methods, and respective components thereof as described herein, which are exclusive of any element not recited in that description of the embodiment.
  • the term “consisting essentially of” refers to those elements required for a given embodiment. The term permits the presence of additional elements that do not materially affect the basic and novel or functional characteristic(s) of that embodiment of the invention.
  • Described herein is the molecular characterization of bronchial premalignant lesions and the airway field of injury identified epithelial and immune alterations associated with progressive/persistent bronchial dysplasia that can be leveraged to develop lung cancer risk biomarkers and interception strategies.
  • Bronchial premalignant lesions are precursors of lung squamous cell carcinoma, but have variable outcome, and tools are lacking to identify and treat PMLs at highest risk for progression to invasive cancer.
  • Profiling endobronchial biopsies of PMLs obtained from high-risk smokers by RNA-Seq identified four PML subtypes with differences in epithelial and immune processes.
  • One molecular subtype (Proliferative) is enriched with dysplastic lesions and exhibits up-regulation of metabolic and cell cycle pathways and down-regulation of ciliary processes.
  • RNA-Seq profiles from normal-appearing uninvolved large airway brushings could identify subjects with Proliferative lesions with high specificity.
  • Lung cancer is the leading cause of cancer death taking about 160,000 U.S. lives each year, more than colorectal, pancreatic, breast, and prostate cancers combined.
  • innovative strategies are needed to intercept cancer development by diagnosing the disease at its earliest and potentially most curable stage.
  • Recent advances based on results from the National Lung Screening Trial (1) are dramatically altering the landscape of early LC detection as computed tomography (CT) screening of high-risk individuals significantly reduces mortality.
  • CT computed tomography
  • biomarkers are needed to select individuals for LC screening as eligibility criteria account for less than 27% of individuals diagnosed with LC in the US (2) and to distinguish between benign or cancerous indeterminate pulmonary nodules as screening has very high false positive rate (>90%).
  • CT computed tomography
  • LC risk biomarkers and LC interception strategies requires a detailed understanding of the earliest molecular alterations involved in lung carcinogenesis that occur in the respiratory epithelium (3, 4). Exposure to cigarette smoke creates a field of injury throughout the entire respiratory tract by inducing a variety of genomic alterations that can lead to an “at-risk” airway where premalignant lesions (PMLs) and LCs develop.
  • PMLs premalignant lesions
  • Lung squamous cell carcinoma arises in the epithelial layer of the bronchial airways and is often preceded by the development of PMLs through a stepwise histological progression from normal epithelium to hyperplasia, squamous metaplasia, dysplasia (mild, moderate and severe), carcinoma in situ (CIS), and finally to invasive and then metastatic LUSC (5).
  • LUSC Lung squamous cell carcinoma
  • bronchial biopsies containing a mixture of epithelial and immune cells would allow us to identify transcriptomic alterations associated with high-grade histology and premalignant lesion progression.
  • mRNA sequencing was used to profile endobronchial biopsies and brushings obtained through serial bronchoscopies from high-risk smokers undergoing lung cancer screening by auto-fluorescence bronchoscopy and chest CT.
  • Using the bronchial biopsies four molecular subtypes associated with clinical phenotypes and biological processes were identified.
  • One subtype is enriched with biopsies having dysplastic histology, high basal cell and low ciliated cell signals, and expression of proliferation-associated pathways. Genes involved in interferon signaling and T cell mediated immunity were down-regulated among progressive/persistent lesions within the Proliferative subtype compared with regressive lesions and these pathways correlated with decreases in both innate and adaptive immune cell types. Molecular classification of biopsies into a high-grade/progressive disease group can be used to stratify patients into prevention trials and to monitor efficacy of the treatment. The results also indicate that personalized lung cancer chemoprevention targeting specific cancer-related pathways or the immune system can have potential therapeutic benefits.
  • mRNA sequencing was used to profile endobronchial biopsies and brushings obtained through serial bronchoscopy of high-risk smokers undergoing lung cancer screening by auto-fluorescence bronchoscopy and chest CT at the Roswell Park Comprehensive Cancer Center (Roswell) in Buffalo, N.Y.
  • the subjects are predominantly older smokers, many of which have a history of lung cancer, chronic obstructive pulmonary disease (COPD), and occupational exposures that confer a high-risk of developing lung cancer.
  • Clinical characteristics reported at baseline such as sex, age, smoking status (ever or never) reported at baseline visit, pack-years, prior history of lung cancer, COPD status, and occupational exposures were not significantly different between the two cohorts (Table 1).
  • the DC had 190 biopsies and 89 brushes while the VC had 105 biopsies and 48 brushes.
  • Ninety-four percent of subjects had at least one lung anatomic location sampled 2 or more times via endobronchial biopsy.
  • the DC and VC contained 37.9% and 35.2% biopsies with a histological grade of dysplasia or higher and 23.1% and 19.0% had progressive/persistent dysplasia, respectively (Table 2).
  • a previously described smoking-associated signature (8) was used to predict the smoking status of each sample, as smoking status was only available at baseline, and found that the DC had a higher percentage of biopsies predicted to be current smokers (62.6%) compared with the VC (36.2%).
  • the predicted smoking status was consistent across all procedures for 63% and 70% of the DC and VC subjects, respectively.
  • the DC had significantly greater total reads, percent uniquely mapping reads, and median transcript integrity number scores among the biopsies than the VC, but these differences between cohorts were not reflected in the brushes ( FIG. 5 ).
  • WGCNA weighted gene co-expression network analysis
  • NTCU n-nitrosotris-(2-choroethyl)urea
  • DC biopsy gene modules that were highly correlated (absolute Pearson correlation coefficient r>0.85) to at least one other non-DC biopsy module within each of the 4 datasets were selected. Genes in the selected modules were filtered by requiring that each gene was also present in at least one of the correlated non-DC biopsy modules, resulting in a set of 9 gene modules that consisted of 3,936 genes in total ( FIG. 6 ).
  • each molecular subtype In order to characterize each molecular subtype, the first focus was on identifying biological pathways over-represented in the genes comprising each gene module, as the pattern of gene module expression defines each PML subtype. Each gene module was found to be associated with distinct epithelial and immune biological processes ( FIG. 1 A , FIG. 6 , and Table 5).
  • the Proliferative subtype is specifically characterized by increased expression of genes involved in energy metabolism and cell cycle pathways (Modules 4 and 5).
  • the Secretory and Normal-like subtypes both have increased expression of genes in cilium-associated pathways (Module 6), however, the Normal-like subtype specifically has decreased expression of genes involved in inflammation, regulation of lymphocytes and leukocytes, and antigen processing and presentation pathways (Modules 8 and 9).
  • the Secretory subtype exhibits decreased expression of genes involved in protein translation (Module 7), while RNA processing genes (Module 2) are expressed more highly in the Inflammatory subtype.
  • the molecular subtypes were further characterized by their associations with clinical phenotypes and established LUSC tumor molecular subtypes (11, 12). Sample smoking status, the subject from whom the sample was derived, and sample histology demonstrated significant associations with subtype (p ⁇ 0.01, FIG. 1 B , Table 6, FIG. 8 ). The Proliferative and Secretory subtypes are enriched for current smokers and this association drives the subject enrichment as 79% of subjects maintain their smoking status throughout the study. Additionally, the Proliferative subtype is enriched for biopsies with dysplasia histology ( FIG. 1 B ).
  • the Proliferative subtype samples also had high concordance with the LUSC-Classical subtype ( FIG. 1 B ).
  • the LUSC-Classical subtype was associated with alterations and overexpression of KEAP1 and NFE2L2 as well as amplification of 3q26 with overexpression of SOX2, TP63 and PIK3CA (11).
  • the LUSC-Classical subtype was found to be associated with increased expression of genes involved in energy metabolism, and our Proliferative subtype is in part defined by high expression of Module 4, which is enriched for genes associated with oxidative phosphorylation and the electron transport chain.
  • the Inflammatory and Secretory PML subtypes demonstrate enrichment for the LUSC-Secretory subtype.
  • the LUSC-Secretory subtype was associated with processes related to the immune response, and the Inflammatory and Secretory PMLs have the highest expression of Module 8 that is enriched for genes in these same pathways.
  • the expression levels of these marker genes agree with cell type deconvolution methods to examine epithelial and immune cell content ( FIG. 10 C- 10 D ).
  • a 22-gene nearest centroid molecular subtype predictor was developed by selecting genes representative of each of the 9 gene modules. The predictor has 84.7% accuracy across DC biopsies (training set, FIG. 2 A and FIG. 11 ) with the following misclassification rates per subtype 5/52 (9.6%) in Proliferative, 7/37 (18.9%) in Inflammatory, 9/61 (14.8%) in Secretory, and 8/40 (20%) in Normal-like.
  • the 22-gene classifier was used to predict the molecular subtype of the 105 VC biopsies ( FIG. 2 B ).
  • the VC subtype predictions were evaluated by examining the concordance of metagene scores for each of the 9 modules (using the full set of genes for each module) between the predicted VC subtypes compared with the DC subtypes.
  • the average behavior of Principal Component 1 (PC1) across the subtypes was highly similar ( FIG. 12 ) with few exceptions (namely, Module 3 that had the fewest genes).
  • the statistical associations between the VC subtypes (via the 22-gene classifier) and clinical and molecular phenotypes across the VC biopsies are analogous to those observed across the DC biopsies ( FIG. 2 C , Table 6, FIG. 8 and FIG. 10 A- 10 H ).
  • the Proliferative subtype is enriched for current smokers, biopsies with dysplasia histology, and the LUSC-Classical tumor subtype ( FIG. 2 C , Table 6).
  • MUC5AC a marker of goblet epithelial cells
  • the Proliferative subtype represents a distinct subtype of PMLs enriched for dysplastic histology expressing metabolic and proliferative pathways.
  • Biopsies classified as the Proliferative subtype may represent a group of PMLs that need close monitoring and intervention. As a result, it was sought to explore whether or not it was possible to predict the presence of Proliferative subtype biopsies using the brushes.
  • the Proliferative subtype is defined by the behavior of Modules 4, 5, 6, and 7 (Table 3), and therefore, the subset of 8 genes (from the 22-gene predictor) that correspond to these Modules was used to predict the presence of the Proliferative subtype across the DC and VC biopsies and brushes.
  • a prediction of the Proliferative subtype in a brush is specific (91% and 92% in the DC and VC biopsies, respectively), but not sensitive (39% and 32% DC and VC biopsies, respectively) at indicating the presence of at least one Proliferative PML detected at the same time point ( FIG. 3 A ).
  • GSVA Gene Set Variation Analysis
  • the genes in Module 9 include a number of genes that encode for proteins involved in interferon signaling as well as antigen processing and presentation (SP100, CHTA, CXCL10, SOCS1, GBP1, GBP4, B2M, TAP1, TAPBP, TRIM 14, TRIM21, TRIM22, STAT1, PML, OAS2, OAS3, MX1, ADAR, ISG15, IFI35, IFIT3, IFI27, PSMB8, PSMB9, BST2, IRF1, IRF9, CD74, PSME1, PSME2, HLA-DQA1/DPA1/DPB1/DRA/DQB2/DRB1/DQB1/DMA/DMB/DOA, HLA-A/B/C/E/F) and include the inhibitory receptor LAG3.
  • CD4 T cells were increased (p ⁇ 0.001 in the concordant set, linear model) and CD8 T cells were decreased (p ⁇ 0.001 in the concordant set) in PMLs that progress/persist.
  • the immunofluorescence results did not reach significance, with the exception of CD163, when just the lesion outcome was used without regard to the Module 9 score.
  • Lung squamous cell carcinoma is the second most common form of lung cancer and arises in the epithelial layer of the bronchial airways. It is often preceded by the development of lung squamous premalignant lesions (PMLs). The presence of dysplastic persistent and or progressive PMLs is a marker of increased risk for LUSC (6). Currently, however, effective tools to identify PMLs at highest risk of progression to invasive carcinoma are lacking (7). The development of markers predictive of disease progression will be important in identifying patients at highest risk for LUSC development and in identifying biological pathways exploitable for LUSC chemoprevention.
  • RNA-Seq bronchial brushes and endobronchial biopsies obtained from subjects undergoing longitudinal lung cancer screening by chest computed tomography (CT) and autofluorescence bronchoscopy.
  • CT chest computed tomography
  • bronchoscopy Four transcriptionally distinct groups of biopsies are identified, one of these labelled Proliferative and found to be associated with high-grade dysplasia.
  • Patients with Proliferative PMLs can also be identified via gene expression measured from cells in the non-involved large airway epithelium. It was further found that persistent/progressive Proliferative PMLs are characterized by decreased expression of genes involved in interferon signaling and antigen processing/presentation pathways.
  • Genomic gains in loci containing SOX2, TP63, EGFR, MYC, CEP3, and CEP5 are also associated with progression of high-grade dysplasia (23).
  • PML histological grade and progression Despite the numerous genomic alterations associated with PML histological grade and progression, a comprehensive PML molecular classification system to complement the pathologic classification of PML is lacking.
  • Use of an unsupervised class discovery approach that led to the identification of four distinct molecular PML subtypes (Proliferative, Inflammatory, Secretory, and Normal-like).
  • the transcriptional patterns differentiating the PML subtypes are robust and a 22-gene panel identified in the Discovery Cohort can be used to distinguish between the different molecular subtypes in an independent Validation Cohort.
  • prior lung cancer history may influence airway gene expression and about two-thirds of the subjects have a prior history of lung cancer
  • we do not detect a significant association between lung cancer history and molecular subtype and there is a similar diversity of molecular subtypes between biopsies collected from subjects with and without a lung cancer history.
  • the Proliferative subtype is enriched with dysplastic PMLs from current smokers and is characterized by up-regulation of metabolic (OXPHOS/ETC/TCA) and cell cycle pathways and down-regulation of cilia-associated pathways.
  • the Inflammatory subtype also shows increased expression of a gene module enriched for genes involved in inflammation and regulation of lymphocytes and leukocytes (Module 8).
  • This gene module is also elevated in Secretory lesions predominated by lesions from current smokers and exhibiting increased expression of goblet cell markers.
  • IL1B is part of this inflammation-related gene module, which is of great interest as the inhibition of IL1B has recently been shown to reduce lung cancer incidence (28).
  • the molecular profiling of PMLs and the identification of gene co-expression modules also provides an opportunity to identify the molecular determinants of subsequent PML progression.
  • One of the nine gene co-expression modules used to define the molecular subtypes was significantly different between biopsies that progress or persist compared to biopsies that regress within the Proliferative subtype in both the DC and VC cohorts.
  • the module contains genes whose expression is decreased in the persistent/progressive biopsies that are involved in interferon signaling and antigen processing and presentation. These gene expression changes were correlated with a decreased abundance of innate and adaptive immune cells via computational prediction.
  • epigenetic therapy specifically DNA methyltransferase inhibitors (42) has been shown to enhance response to immune checkpoint therapy and up-regulate many of the genes down-regulated in progressive/persistent lesions within the Proliferative subtype including HLA class I genes (HLA-B and HLA-C), B2M, CD58, TAP1, immune-proteasome subunits PSMB9 and PSMB8, and the transcription factor IRF9.
  • HLA class I genes HLA-B and HLA-C
  • B2M B2M
  • CD58 CD58
  • TAP1 immune-proteasome subunits
  • PSMB9 and PSMB8 the transcription factor IRF9.
  • Unraveling the mechanisms of innate and adaptive immune down-regulation in this subset of PMLs will be important to identifying potential immunoprevention therapies.
  • biomarkers for determining PML subtype and assessing immune infiltration may have utility for the detection of aggressive PMLs that require more intensive clinical management and genes altered in these PMLs may serve as lung chemoprevention candidates.
  • biomarkers could either be measured directly in PML tissue, or as indicated by the present data, they can be measured in a surrogate tissue such as bronchial airway epithelium.
  • a benefit of biomarkers predicting aggressive PML behavior measured in surrogate tissue is the potential that these biomarkers can also predict the behavior of PMLs not directly observed during bronchoscopy.
  • Endobronchial biopsies and brushings were obtained from high-risk subjects undergoing lung cancer screening at approximately 1-year intervals by white light and auto-fluorescence bronchoscopy and computed tomography at Roswell.
  • the bronchoscopy included visualization of the vocal cords, trachea, main carina, and orifices of the sub-segmental bronchi visible without causing trauma to the bronchial wall. All abnormal and suspicious areas are biopsied twice and the lung anatomic location is recorded ( FIG. 14 , Table 8).
  • One biopsy was used for routine pathological evaluation and the other for molecular profiling.
  • a brushing was obtained from a normal appearing area of the left or right mainstem bronchus for research.
  • Eligibility for screening includes either a previous history of aerodigestive cancer and no disease at the time of enrollment or age greater than 50, a current or previous history of smoking for a minimum exposure of 20 pack-years and at least one additional risk factor including moderate chronic obstructive pulmonary disease (COPD) (defined as forced expiratory volume (FEV1) ⁇ 70%), confirmed asbestos related lung disease or a strong family history of lung cancer (at least 1-2 first degree relatives). All research specimens were stored in RNA Allprotect (Qiagen) and stored at ⁇ 80 degrees C.
  • COPD chronic obstructive pulmonary disease
  • FEV1 forced expiratory volume
  • DC discovery cohort
  • VC validation cohort
  • Biopsy progression/regression was defined for each biopsy based on the histology of the biopsy and the worst histology recorded for the same lung anatomic location in the future. Histology changes between normal, hyperplasia, and metaplasia were classified as “normal stable”, decreases in histological dysplasia grade or changes from dysplastic histology to normal/hyperplasia/metaplasia were classified as “regressive”, lack of future histological data was classified as “unknown”, and everything else was classified as “progressive/persistent.” The Institutional Review Boards at Boston University Medical Center and Roswell approved the study and all subjects provided written informed consent.
  • Samples were excluded based on values greater than 2 standard deviations from the mean for more than one of the following criteria: 1) mean Pearson correlation with all other samples calculated across all filtered genes 2) the 1 st or 2 nd principal components calculated using the filtered gene expression matrix 3) transcript integrity number (TIN, computed by RSeQC). After sample filtering, gene filtering was recomputed as described above on the final set of high-quality samples.
  • the data are available from NCBI's Gene Expression Omnibus using the accession GSE109743.
  • LUSC squamous cell carcinoma
  • NTCU n-nitrosotris-(2-choroethyl)urea
  • mice develop lesions that are histologically and molecularly comparable to human lesions and that progress to LUSC and the samples represent a range of histology (normal, mild dysplasia, moderate dysplasia, severe dysplasia, carcinoma in situ (CIS), and LUSC tumor).
  • the mouse data are available from NCBI's Gene Expression Omnibus using the accession ID GSE111091. Sample and gene filtering from the TCGA LUSC tumors and the mouse tissue were processed as described elsewhere herein.
  • WGCNA Weighted correlation network analysis
  • the first principal component (PC1) was calculated across each z-score normalized dataset.
  • PC1 principal component
  • the genes defining the retained biopsy Modules were required to be present in the biopsy Module and at least in one of the correlated gene sets.
  • the residual gene expression values across the reduced set of genes for the discovery biopsies was used as input for consensus clustering (49).
  • Consensus clustering was performed setting k (number of groups) to 10, the number of iterations to 1000, the subsampling to 80%, the clustering algorithm to partitioning around mediods, and the distance metric to Pearson correlation.
  • the optimal value for k was 4 based on the relative change in area under the cumulative distribution function calculated based on the consensus matrix for each k.
  • the DC biopsies across the filtered genes were used to derive a molecular subtype predictor.
  • Pearson correlation metrics were determined between each gene and the Module eigengenes (PC1 for each of the 9 Modules). Genes were retained as part of a Module if the correlation value was the highest for the Module in which it was assigned. The average Pearson correlation of the retained genes to the Module eigengene was computed, and the number of genes chosen from each Module for the predictor was inversely proportional to this metric.
  • the genes most highly correlated to the Module eigengene were chosen to represent the Module in the predictor.
  • the 22 genes resulting from this analysis across the DC biopsy data were used to train a nearest centroid predictor using the pamr package with a threshold of zero and predict the molecular subtype across the VC biopsies. Prior to predicting the molecular subtype of these test sets, the training and test sets were combat (50) adjusted and z-score normalized across combined training and test data. Using the methods described above we derived molecular subtypes using consensus clustering across the VC biopsies and compared these to the predicted subtypes.
  • each gene Module was evaluated by testing if GSVA (14) scores for each Module were significantly (FDR ⁇ 0.05) associated with the molecular subtypes using a linear mixed effect model with patient as a random effect via limma.
  • the molecular subtypes in the DC biopsies were annotated according to the behavior of each gene Module by calculating whether or not GSVA (14) scores for each Module were significantly up- or down-regulated (FDR ⁇ 0.05) in a particular molecular subtype versus all other samples using a linear mixed effects model with patient as a random effect via limma. Additionally, the biological pathways and transcription factors associated with each subtype were identified using GSEA (52) and mSigDB (53) gene sets using genes ranked by the t-statistic for their association with each subtype. The ranked lists were created using the limma (54) and edgeR (48) packages to identify differentially expressed genes associated with subtype membership.
  • Each linear model used voom-transformed (55) data and included membership in the subtype of interest, batch, and RNA quality (TIN) as covariates and patient as a random effect.
  • Pathways enriched in the ranked lists were used to annotate the molecular subtypes. FDR values for individual genes were derived from this analysis or analogous models using only samples of normal/hyperplasia histology or dysplasia histology.
  • a linear mixed effects model was used to assess module GSVA score differences between progressive/persistent versus regressive lesions within each molecular subtype with patient as a random effect via limma.
  • epithelial cell content ‘Epithelial’ in xCell and ‘Normal mucosa’ in Bindea et al.
  • Sensitivity and specificity performance metrics were calculated based on the ability of a Proliferative subtype prediction in the DC or VC brushes to indicate the presence of at least one biopsy of the Proliferative subtype.
  • the behavior of the modules that define the Proliferative subtype in the DC biopsies was analyzed across the DC and VC brushes.
  • Imaging was performed using an Aperio Slide Scanner for scoring and a Carl Zeiss Axio (20x and 40 x objectives) and a Carl Zeiss LSM 710 NLO confocal microscope for capturing additional images.
  • Digital slides were analyzed with the Definiens Tissue Studio (Definiens Inc.) for the enumeration of immunofluorescence staining.
  • the enumeration of the immunofluorescence scored each stain including DAPI positive cells.
  • the enumeration was conducted on different regions (independent areas of tissue) present on a slide (1-5 regions/biopsy) for each biopsy. For each region, the percentage of positively staining cells for a given protein was calculated by dividing the number of positively stained cells by the total number of DAPI positive cells.
  • a binomial mixed effects model via the lme4 R package was used to assess differences in the percentages of cells staining positive for a given protein in each region between progressive/persistent versus regressive biopsies using the total cells stained in each region as weights and adjusting for the slide number as a random effect.
  • the models were used across samples from the Proliferative subtype and across samples from the Proliferative subtype where the biopsy outcome (progressive/persistent versus regressive) agreed with the Module 9 GSVA score (scores less than 0 are associated with progression/persistence and scores greater than 0 are associated with regression).
  • Each region was also qualitatively scored as either positive or negative for having a distinct CD8 T cell localization pattern where cells lined and were embedded within the epithelium.
  • N-nitrosotris-(2-choroethyl)urea mouse sample collection and library preparation.
  • RNA was collected and banked RNA from 40 fresh frozen whole lung sections (curls) and laser microdissected (LCM) tissue isolated with an Acrutus PixcellTM II, from SWR/J and A/J mice treated with NTCU.
  • Mice had been treated topically with 15 or 25 umol NTCU (25 ul of 40 mM NTCU for 15 or 25 weeks) as part of a study performed in accordance with IACUC approved protocol at RPCC.
  • Sequencing libraries will be prepared from total RNA samples using Illumina® TruSeq® RNA Sample Preparation Kit v2. Each sample was sequenced five per lane on the Illumina® HiSeq 2500 to generate single-end 50-nucleotide reads.
  • NTCU mouse data processing Demultiplexing and creation of FASTQ files were performed using Illumina CASAVA 1.8.2. Trimmomatic was used to trim adapter sequences as well as to trim reads of poor quality using the following parameters: ILLUMINACLIP:TruSeq3-SE.fa:2:30:10, LEADING:20, TRAILING:20, SLIDINGWINDOW:4:20, and MINLEN:20. After trimming, greater than 99% of reads were retained in all samples. Samples were subsequently aligned using mm9 and 2-pass STAR(44) alignment. Gene and transcript level counts were calculated using RSEM(45) using Ensembl annotation. Quality metrics were calculated by STAR and RSeQC(46).
  • a binomial mixed effects model via the lme4 R package was used to assess differences in the percentages of cells staining positive for a given protein in each region between the molecular subtypes using the total cells stained in each region as weights and adjusting for patient as a random effect.
  • Table 5 depicts pathways enriched in the Gene Modules. Enrichr results (FDR ⁇ 0.05) for selected pathways associated with each gene modules.
  • Previous history of lung cancer was categorized as follows: no history (No LC History), a previous history of LC that include a lung squamous cell carcinoma (LC History - LUSC), and a previous history of LC that does not include a lung squamous cell carcinoma (LC History - Other).
  • No LC History a previous history of LC that include a lung squamous cell carcinoma
  • LC History - Other a previous history of LC that does not include a lung squamous cell carcinoma

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • Physics & Mathematics (AREA)
  • Molecular Biology (AREA)
  • Medical Informatics (AREA)
  • Public Health (AREA)
  • Pathology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Analytical Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Epidemiology (AREA)
  • Data Mining & Analysis (AREA)
  • Biophysics (AREA)
  • Food Science & Technology (AREA)
  • General Physics & Mathematics (AREA)
  • Cell Biology (AREA)
  • Databases & Information Systems (AREA)
  • Oncology (AREA)
  • Hospice & Palliative Care (AREA)
  • Primary Health Care (AREA)
  • Theoretical Computer Science (AREA)
  • Spectroscopy & Molecular Physics (AREA)
  • Evolutionary Biology (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Pharmacology & Pharmacy (AREA)
US16/545,032 2018-08-20 2019-08-20 Methods related to bronchial premalignant lesion severity and progression Active US11579140B2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US16/545,032 US11579140B2 (en) 2018-08-20 2019-08-20 Methods related to bronchial premalignant lesion severity and progression
US18/093,066 US20230146253A1 (en) 2018-08-20 2023-01-04 Methods related to bronchial premalignant lesion severity and progression

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201862765264P 2018-08-20 2018-08-20
US16/545,032 US11579140B2 (en) 2018-08-20 2019-08-20 Methods related to bronchial premalignant lesion severity and progression

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US18/093,066 Division US20230146253A1 (en) 2018-08-20 2023-01-04 Methods related to bronchial premalignant lesion severity and progression

Publications (2)

Publication Number Publication Date
US20200057053A1 US20200057053A1 (en) 2020-02-20
US11579140B2 true US11579140B2 (en) 2023-02-14

Family

ID=69523934

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/545,032 Active US11579140B2 (en) 2018-08-20 2019-08-20 Methods related to bronchial premalignant lesion severity and progression
US18/093,066 Pending US20230146253A1 (en) 2018-08-20 2023-01-04 Methods related to bronchial premalignant lesion severity and progression

Family Applications After (1)

Application Number Title Priority Date Filing Date
US18/093,066 Pending US20230146253A1 (en) 2018-08-20 2023-01-04 Methods related to bronchial premalignant lesion severity and progression

Country Status (11)

Country Link
US (2) US11579140B2 (ko)
EP (1) EP3840634A4 (ko)
JP (1) JP2021536434A (ko)
KR (1) KR20210049117A (ko)
CN (1) CN112867435A (ko)
AU (1) AU2019326405A1 (ko)
BR (1) BR112021003095A2 (ko)
CA (1) CA3109391A1 (ko)
IL (1) IL280815A (ko)
MX (1) MX2021002071A (ko)
WO (1) WO2020041243A1 (ko)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111876478A (zh) * 2020-04-28 2020-11-03 中国科学院微生物研究所 肺结节诊断标志物及应用
CN113109563B (zh) * 2020-05-27 2022-05-06 郑州大学第一附属医院 用于高危人群早期食管鳞癌筛查的标志物及其应用
CN111920961B (zh) * 2020-08-14 2023-09-08 福建医科大学附属协和医院 一种治疗癌症的药物
CN114544955B (zh) * 2020-11-26 2023-09-15 四川大学华西医院 Gasp-2检测试剂在制备肺癌早期诊断和易感性检测试剂盒中的用途
US11946928B2 (en) * 2021-05-20 2024-04-02 Trustees Of Boston University Methods and compositions relating to airway dysfunction
CN113151482A (zh) * 2021-05-21 2021-07-23 深圳泰莱生物科技有限公司 一种基于单色多重荧光定量pcr的鉴别良恶性肺结节的方法
CN115364231B (zh) * 2021-10-15 2023-11-17 北京大学第三医院(北京大学第三临床医学院) 一种增强ezh2抑制剂抗肿瘤作用的药物组合物及其用途
CN114292919A (zh) * 2022-01-11 2022-04-08 广东省人民医院 一种早期Luminal乳腺癌风险诊断试剂盒

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060165773A1 (en) 1998-01-06 2006-07-27 Roman Perez-Soler Gene therapy of tumors using non-viral delivery system
US20100055047A1 (en) 2008-08-26 2010-03-04 Yiyu Zou Methods for treating bronchial premalignancy and lung cancer
US20180010197A1 (en) 2016-07-08 2018-01-11 Trustees Of Boston University Gene expression-based biomarker for the detection and monitoring of bronchial premalignant lesions
US20180171418A1 (en) 2005-04-14 2018-06-21 Trustees Of Boston University Diagnostic for lung disorders using class prediction
WO2018136919A2 (en) 2017-01-23 2018-07-26 Trustees Of Boston University Methods relating to lung cancer

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060165773A1 (en) 1998-01-06 2006-07-27 Roman Perez-Soler Gene therapy of tumors using non-viral delivery system
US20180171418A1 (en) 2005-04-14 2018-06-21 Trustees Of Boston University Diagnostic for lung disorders using class prediction
US20100055047A1 (en) 2008-08-26 2010-03-04 Yiyu Zou Methods for treating bronchial premalignancy and lung cancer
US20180010197A1 (en) 2016-07-08 2018-01-11 Trustees Of Boston University Gene expression-based biomarker for the detection and monitoring of bronchial premalignant lesions
WO2018136919A2 (en) 2017-01-23 2018-07-26 Trustees Of Boston University Methods relating to lung cancer

Non-Patent Citations (14)

* Cited by examiner, † Cited by third party
Title
Alaa, M. et al. Lung Cancer 72:303-308. (Year: 2011). *
Beane et al. "Detecting the Presence and Progression of Premalignant Lung Lesions via Airway Gene Expression." Clin. Cancer Res. 23(17): 5091-5100 (2017).
Gao et al. "Loss of IFN-gamma Pathway Genes in Tumor Cells as a Mechanism of Resistance to Anti-CTLA-4 Therapy." Cell 167(2): 397-404.e9 (2016).
Gettinger et al. "Impaired HLA Class I Antigen Processing and Presentation as a Mechanism of Acquired Resistance to Immune Checkpoint Inhibitors in Lung Cancer." Cancer Discov 7(12): 1420-1435 (2017).
Gustafson et al. "Airway PI3K pathway activation is an early and reversible event in lung cancer development." Science translational medicine 2.26 (2010): 26ra25-26ra25.
Kotsakis et al. "Prognostic value of circulating regulatory T cell subsets in untreated non-small cell lung cancer patients." Sci Rep 6: 39247 p. 1-11 (2016).
Liu, X. et al. International Journal of Oncology 45:1266 (2014). (Year: 2014). *
Lonergan, K. et al. PLoS One 5(2):e9162 (2010). (Year: 2010). *
Pereira et al. "Genomic Profiling of Patient-Derived Xenografts for Lung Cancer Identifies B2M Inactivation Impairing Immunorecognition." Clin. Cancer Res. 23(12): 3203-3213 (2017).
Schneider, M. et al. International Journal of Oncology 50:365 (Feb. 2017). (Year: 2017). *
Specht, E. et al. Oncology Research and Treatment 38 (Suppl 5):abstract P461 (Oct. 2015). (Year: 2015). *
Wang et al. "Expression of CD163, interleukin-10, and interferon-gamma in oral squamous cell carcinoma: mutual relationships and prognostic implications." Eur. J. Oral Sci. 122(3): 202-209 (2014).
Wikman, H. et al. Oncogene 21:5804-5813. (Year: 2002). *
Wu et al. "Stromal PD-L1-Positive Regulatory T cells and PD-1-Positive CD8-Positive T cells Define the Response of Different Subsets of Non-Small Cell Lung Cancer to PD-1/PD-L1 Blockade Immunotherapy." J. Thorac Oncol 13(4): 521-532 (2018).

Also Published As

Publication number Publication date
US20230146253A1 (en) 2023-05-11
IL280815A (en) 2021-04-29
BR112021003095A2 (pt) 2021-05-11
JP2021536434A (ja) 2021-12-27
MX2021002071A (es) 2021-04-28
EP3840634A4 (en) 2022-05-11
US20200057053A1 (en) 2020-02-20
WO2020041243A1 (en) 2020-02-27
WO2020041243A8 (en) 2020-04-16
KR20210049117A (ko) 2021-05-04
EP3840634A1 (en) 2021-06-30
AU2019326405A1 (en) 2021-02-18
CN112867435A (zh) 2021-05-28
CA3109391A1 (en) 2020-02-27

Similar Documents

Publication Publication Date Title
US11579140B2 (en) Methods related to bronchial premalignant lesion severity and progression
US20210222254A1 (en) Methods for subtyping of lung adenocarcinoma
EP3430171B1 (en) Detection and treatment of anti-pd-1 therapy resistant metastatic melanomas
EP1934377B1 (en) Methods for identifying biomarkers useful in diagnosis of biological states
EP2653546B1 (en) Marker for predicting stomach cancer prognosis and method for predicting stomach cancer prognosis
EP2925885B1 (en) Molecular diagnostic test for cancer
US11041214B2 (en) Methods for subtyping of lung squamous cell carcinoma
WO2019070755A1 (en) METHODS AND COMPOSITIONS FOR DETECTING AND MODULATING A GENETIC SIGNATURE OF IMMUNOTHERAPY RESISTANCE IN CANCER
US20170283884A1 (en) Methods for predicting drug responsiveness
EP2944961A1 (en) Markers for cancer prognosis and therapy and methods of use
US20130303826A1 (en) Prognostic signature for oral squamous cell carcinoma
US20190367964A1 (en) Dissociation of human tumor to single cell suspension followed by biological analysis
US20210164056A1 (en) Use of metastases-specific signatures for treatment of cancer
US20220025465A1 (en) Systems and methods for characterizing and treating cancer
US20140308241A1 (en) Biomarkers for t cell malignancies and uses thereof
US10969390B2 (en) Method for evaluating efficacy of chemoradiotherapy against squamous cell carcinoma
WO2014072086A1 (en) Biomarkers for prognosis of lung cancer
JP2022536075A (ja) 処置レジメンを調節する方法
US20230257825A1 (en) Breast cancer biomarkers and methods of use

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

AS Assignment

Owner name: TRUSTEES OF BOSTON UNIVERSITY, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BEANE-EBEL, JENNIFER E.;SPIRA, AVRUM E.;LENBURG, MARC;AND OTHERS;SIGNING DATES FROM 20190905 TO 20190916;REEL/FRAME:050409/0949

AS Assignment

Owner name: HEALTH RESEARCH, INC., NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:REID, MARY E.;REEL/FRAME:051251/0244

Effective date: 20191209

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE AFTER FINAL ACTION FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: ADVISORY ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: ADVISORY ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STPP Information on status: patent application and granting procedure in general

Free format text: AWAITING TC RESP., ISSUE FEE NOT PAID

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STCF Information on status: patent grant

Free format text: PATENTED CASE